1
|
Wu J, Li R, Wang J, Zhu H, Ma Y, You C, Shu K. Reactive Astrocytes in Glioma: Emerging Opportunities and Challenges. Int J Mol Sci 2025; 26:2907. [PMID: 40243478 PMCID: PMC11989224 DOI: 10.3390/ijms26072907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/16/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Gliomas are the most prevalent malignant tumors in the adult central nervous system (CNS). Glioblastoma (GBM) accounts for approximately 60-70% of primary gliomas. It is a great challenge to human health because of its high degree of malignancy, rapid progression, short survival time, and susceptibility to recurrence. Owing to the specificity of the CNS, the glioma microenvironment often contains numerous glial cells. Astrocytes are most widely distributed in the human brain and form reactive astrocyte proliferation regions around glioma tissue. In addition, astrocytes are activated under pathological conditions and regulate tumor and microenvironmental cells through cell-to-cell contact or the secretion of active substances. Therefore, astrocytes have attracted attention as important components of the glioma microenvironment. Here, we focus on the mechanisms of reactive astrocyte activation under glioma conditions, their contribution to the mechanisms of glioma genesis and progression, and their potential value as targets for clinical intervention in gliomas.
Collapse
Affiliation(s)
| | | | | | | | | | - Chao You
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095, Jie Fang Avenue, Qiao Kou District, Wuhan 430030, China; (J.W.); (J.W.); (H.Z.); (Y.M.)
| | - Kai Shu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095, Jie Fang Avenue, Qiao Kou District, Wuhan 430030, China; (J.W.); (J.W.); (H.Z.); (Y.M.)
| |
Collapse
|
2
|
Liu S, Du X, Chen Z, Zhou R, Wang H, Mao X, Du J, Zhang G, Li H, Song Y, Chang L, Wu Y. Activation of astrocytic NMDA receptors counteracted Aβ-induced reduction of BDNF and elevation of GFAP and complement 3 in the hippocampal astrocytes. Neuroscience 2024; 559:303-315. [PMID: 39276842 DOI: 10.1016/j.neuroscience.2024.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/20/2024] [Accepted: 09/09/2024] [Indexed: 09/17/2024]
Abstract
N-methyl-D-aspartate receptors (NMDARs) play a crucial role in mediating Amyloid-β (Aβ) synaptotoxicity. Our previous studies have demonstrated an opposite (neuroprotection and neurotoxicity) effect of activating astrocytic and neuronal NMDARs with higher dose (10 μM) of NMDA, an agonist of NMDARs. By contrast, activating neuronal or astrocyitc NMDARs with lower dose (1 μM) of NMDA both exerts neuroprotective effect in Aβ-induced neurotoxicity. However, the underlying mechanism of activating astrocytic NMDARs with lower dose of NMDA to protect against Aβ neurotoxicity remains unclear. Based on our previous related work, in this study, using a co-cultured cell model of primary hippocampal neurons and astrocytes, we further investigated the possible factors involved in 1 μM of NMDA activating astrocytic NMDARs to oppose Aβ-induced synaptotoxicity. Our results showed that activation of astrocytic NMDARs by 1 μM NMDA rescued Aβ-induced reduction of brain-derived neurotrophic factor (BDNF), and inhibited Aβ-induced increase of GFAP, complement 3 (C3) and activation of NF-κB. Furthermore, blockade of astrocytic GluN2A with TCN201 abrogated the ability of 1 μM NMDA to counteract the effects of Aβ decreasing BDNF, and increasing GFAP, C3 and activation of NF-κB. These findings suggest that activation of astrocytic NMDARs protect against Aβ-induced synaptotoxicity probably through elevating BDNF and suppressing GFAP and C3. Our present research provides valuable insights for elucidating the underlying mechanism of astrocytic NMDARs activation resisting the toxic effects of Aβ.
Collapse
Affiliation(s)
- Siyu Liu
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Xiaoqiang Du
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Ziyan Chen
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Ruying Zhou
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Hongqi Wang
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Xin Mao
- Department of Radiology, Peking University Third Hospital, Beijing 100068, China
| | - Jiahe Du
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Guitao Zhang
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Hui Li
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Yizhi Song
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Lirong Chang
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China.
| | - Yan Wu
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China; College of Veterinary Medicine, Beijing University of Agriculture, Beijing 100096, China.
| |
Collapse
|
3
|
Cui Y, Rolova T, Fagerholm SC. The role of integrins in brain health and neurodegenerative diseases. Eur J Cell Biol 2024; 103:151441. [PMID: 39002282 DOI: 10.1016/j.ejcb.2024.151441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/15/2024] Open
Abstract
Integrins are heterodimeric membrane proteins expressed on the surface of most cells. They mediate adhesion and signaling processes relevant for a wealth of physiological processes, including nervous system development and function. Interestingly, integrins are also recognized therapeutic targets for inflammatory diseases, such as multiple sclerosis. Here, we discuss the role of integrins in brain development and function, as well as in neurodegenerative diseases affecting the brain (Alzheimer's disease, multiple sclerosis, stroke). Furthermore, we discuss therapeutic targeting of these adhesion receptors in inflammatory diseases of the brain.
Collapse
Affiliation(s)
- Yunhao Cui
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki 00790, Finland
| | - Taisia Rolova
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki 00290, Finland
| | - Susanna C Fagerholm
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki 00790, Finland.
| |
Collapse
|
4
|
Manning D, Rivera EJ, Santana LF. The life cycle of a capillary: Mechanisms of angiogenesis and rarefaction in microvascular physiology and pathologies. Vascul Pharmacol 2024; 156:107393. [PMID: 38857638 PMCID: PMC12051481 DOI: 10.1016/j.vph.2024.107393] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/02/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024]
Abstract
Capillaries are the smallest blood vessels (<10 μm in diameter) in the body and their walls are lined by endothelial cells. These microvessels play a crucial role in nutrient and gas exchange between blood and tissues. Capillary endothelial cells also produce vasoactive molecules and initiate the electrical signals that underlie functional hyperemia and neurovascular coupling. Accordingly, capillary function and density are critical for all cell types to match blood flow to cellular activity. This begins with the process of angiogenesis, when new capillary blood vessels emerge from pre-existing vessels, and ends with rarefaction, the loss of these microvascular structures. This review explores the mechanisms behind these processes, emphasizing their roles in various microvascular diseases and their impact on surrounding cells in health and disease. We discuss recent work on the mechanisms controlling endothelial cell proliferation, migration, and tube formation that underlie angiogenesis under physiological and pathological conditions. The mechanisms underlying functional and anatomical rarefaction and the role of pericytes in this process are also discussed. Based on this work, a model is proposed in which the balance of angiogenic and rarefaction signaling pathways in a particular tissue match microvascular density to the metabolic demands of the surrounding cells. This negative feedback loop becomes disrupted during microvascular rarefaction: angiogenic mechanisms are blunted, reactive oxygen species accumulate, capillary function declines and eventually, capillaries disappear. This, we propose, forms the foundation of the reciprocal relationship between vascular density, blood flow, and metabolic needs and functionality of nearby cells.
Collapse
Affiliation(s)
- Declan Manning
- Department of Physiology & Membrane Biology, School of Medicine, University of California, Davis, United States of America.
| | - Ernesto J Rivera
- Department of Physiology & Membrane Biology, School of Medicine, University of California, Davis, United States of America
| | - L Fernando Santana
- Department of Physiology & Membrane Biology, School of Medicine, University of California, Davis, United States of America
| |
Collapse
|
5
|
Yu H, Liu Y, Xu F, Fu Y, Yang M, Ding L, Wu Y, Tang F, Qiao J, Wen L. A human fetal cerebellar map of the late second trimester reveals developmental molecular characteristics and abnormality in trisomy 21. Cell Rep 2024; 43:114586. [PMID: 39137113 DOI: 10.1016/j.celrep.2024.114586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/29/2024] [Accepted: 07/19/2024] [Indexed: 08/15/2024] Open
Abstract
Our understanding of human fetal cerebellum development during the late second trimester, a critical period for the generation of astrocytes, oligodendrocytes, and unipolar brush cells (UBCs), remains limited. Here, we performed single-cell RNA sequencing (scRNA-seq) in human fetal cerebellum samples from gestational weeks (GWs) 18-25. We find that proliferating UBC progenitors distribute in the subventricular zone of the rhombic lip (RLSVZ) near white matter (WM), forming a layer structure. We also delineate two trajectories from astrogenic radial glia (ARGs) to Bergmann glial progenitors (BGPs) and recognize oligodendrogenic radial glia (ORGs) as one source of primitive oligodendrocyte progenitor cells (PriOPCs). Additionally, our scRNA-seq analysis of the trisomy 21 fetal cerebellum at this stage reveals abnormal upregulated genes in pathways such as the cell adhesion pathway and focal adhesion pathway, which potentially promote neuronal differentiation. Overall, our research provides valuable insights into normal and abnormal development of the human fetal cerebellum.
Collapse
Affiliation(s)
- Hongmin Yu
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing 100871, China
| | - Yun Liu
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing 100871, China; Changping Laboratory, Changping Laboratory, Yard 28, Science Park Road, Changping District, Beijing 102206, China
| | - Fanqing Xu
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
| | - Yuanyuan Fu
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing 100871, China
| | - Ming Yang
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
| | - Ling Ding
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
| | - Yixuan Wu
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
| | - Fuchou Tang
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing 100871, China; Changping Laboratory, Changping Laboratory, Yard 28, Science Park Road, Changping District, Beijing 102206, China
| | - Jie Qiao
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China.
| | - Lu Wen
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing 100871, China; Changping Laboratory, Changping Laboratory, Yard 28, Science Park Road, Changping District, Beijing 102206, China.
| |
Collapse
|
6
|
Ahuja S, Lazar IM. Proteomic insights into breast cancer response to brain cell-secreted factors. Sci Rep 2024; 14:19351. [PMID: 39169222 PMCID: PMC11339284 DOI: 10.1038/s41598-024-70386-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024] Open
Abstract
The most devastating feature of cancer cells is their ability to metastasize to distant sites in the body. HER2 + and TN breast cancers frequently metastasize to the brain and stay potentially dormant for years until favorable conditions support their proliferation. The sheltered and delicate nature of the brain prevents, however, early disease detection and effective delivery of therapeutic drugs. Moreover, the challenges associated with the acquisition of brain biopsies add compounding difficulties to exploring the mechanistic aspects of tumor development. To provide insights into the determinants of cancer cell behavior at the brain metastatic site, this study was aimed at exploring the early response of HER2 + breast cancer cells (SKBR3) to factors present in the brain perivascular niche. The neural microenvironment was simulated by using the secretome of a set of brain cells that come first in contact with the cancer cells upon crossing the blood brain barrier, i.e., endothelial cells, astrocytes, and microglia. Cytokine microarrays were used to investigate the secretome mediators of intercellular communication, and proteomic technologies for assessing the changes in the behavior of cancer cells upon exposure to the brain cell-secreted factors. The cytokines detected in the brain secretomes were supportive of inflammatory conditions, while the SKBR3 cells secreted numerous cancer-promoting growth factors that were either absent or present in lower abundance in the brain cell cultures, indicating that upon exposure the SKBR3 cells may have been deprived of favorable conditions for optimal growth. Altogether, the results suggest that the exposure of SKBR3 cells to the brain cell-secreted factors altered their growth potential and drove them toward a state of quiescence, with broader overall outcomes that affected cellular metabolism, adhesion and immune response processes. The findings of this study underscore the key role played by the neural niche in shaping the behavior of metastasized cancer cells, provide insights into the cellular cross-talk that may lead cancer cells into dormancy, and highlight novel opportunities for the development of metastatic breast cancer therapeutic strategies.
Collapse
Affiliation(s)
- Shreya Ahuja
- Department of Biological Sciences, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA, 24061, USA
| | - Iulia M Lazar
- Department of Biological Sciences, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA, 24061, USA.
- Fralin Life Sciences Institute, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA, 24061, USA.
- Carilion School of Medicine, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA, 24061, USA.
- Division of Systems Biology/AIS, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA, 24061, USA.
| |
Collapse
|
7
|
Guo Y, Zhang S, Wang D, Heng BC, Deng X. Role of cell rearrangement and related signaling pathways in the dynamic process of tip cell selection. Cell Commun Signal 2024; 22:24. [PMID: 38195565 PMCID: PMC10777628 DOI: 10.1186/s12964-023-01364-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 10/25/2023] [Indexed: 01/11/2024] Open
Abstract
Angiogenesis is a complex, highly-coordinated and multi-step process of new blood vessel formation from pre-existing blood vessels. When initiated, the sprouting process is spearheaded by the specialized endothelial cells (ECs) known as tip cells, which guide the organization of accompanying stalk cells and determine the function and morphology of the finally-formed blood vessels. Recent studies indicate that the orchestration and coordination of angiogenesis involve dynamic tip cell selection, which is the competitive selection of cells to lead the angiogenic sprouts. Therefore, this review attempt to summarize the underlying mechanisms involved in tip cell specification in a dynamic manner to enable readers to gain a systemic and overall understanding of tip cell formation, involving cooperative interaction of cell rearrangement with Notch and YAP/TAZ signaling. Various mechanical and chemical signaling cues are integrated to ensure the right number of cells at the right place during angiogenesis, thereby precisely orchestrating morphogenic functions that ensure correct patterning of blood vessels. Video Abstract.
Collapse
Affiliation(s)
- Yaru Guo
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Shihan Zhang
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Dandan Wang
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Boon Chin Heng
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
- NMPA Key Laboratory for Dental Materials, Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
| | - Xuliang Deng
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China.
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China.
- Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
| |
Collapse
|
8
|
Ahuja S, Lazar IM. Proteomic Insights into Metastatic Breast Cancer Response to Brain Cell-Secreted Factors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.22.563488. [PMID: 37961261 PMCID: PMC10634729 DOI: 10.1101/2023.10.22.563488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The most devastating feature of cancer cells is their ability to metastasize to distant sites in the body. HER2+ and triple negative breast cancers frequently metastasize to the brain and stay potentially dormant for years, clinging to the microvasculature, until favorable environmental conditions support their proliferation. The sheltered and delicate nature of the brain prevents, however, early disease detection, diagnosis, and effective delivery of therapeutic drugs. Moreover, the challenges associated with the acquisition of brain tissues and biopsies add compounding difficulties to exploring the mechanistic aspects of tumor development, leading to slow progress in understanding the drivers of disease progression and response to therapy. To provide insights into the determinants of cancer cell behavior at the brain metastatic site, this study was aimed at exploring the growth and initial response of HER2+ breast cancer cells (SKBR3) to factors present in the brain perivascular niche. The neural microenvironment conditions were simulated by using the secretome of a set of brain cells that come first in contact with the cancer cells upon crossing the blood brain barrier, i.e., human endothelial cells (HBEC5i), human astrocytes (NHA) and human microglia (HMC3) cells. Cytokine microarrays were used to investigate the cell secretomes and explore the mediators responsible for cell-cell communication, and proteomic technologies for assessing the changes in the behavior of cancer cells upon exposure to the brain cell-secreted factors. The results of the study suggest that the exposure of SKBR3 cells to the brain secretomes altered their growth potential and drove them towards a state of quiescence. The cytokines, growth factors and enzymes detected in the brain cell-conditioned medium were supportive of mostly inflammatory conditions, indicating a collective functional contribution to cell activation, defense, inflammatory responses, chemotaxis, adhesion, angiogenesis, and ECM organization. The SKBR3 cells, on the other hand, secreted numerous cancer-promoting growth factors that were either absent or present in lower abundance in the brain cell culture media, suggesting that upon exposure the SKBR3 cells were deprived of favorable environmental conditions required for optimal growth. The findings of this study underscore the key role played by the neural niche in shaping the behavior of metastasized cancer cells, providing insights into the cancer-host cell cross-talk that contributes to driving metastasized cancer cells into dormancy and into the opportunities that exist for developing novel therapeutic strategies that target the brain metastases of breast cancer.
Collapse
Affiliation(s)
- Shreya Ahuja
- Department of Biological Sciences, Virginia Tech 1981 Kraft Drive, Blacksburg, VA 24061
| | - Iulia M. Lazar
- Department of Biological Sciences, Virginia Tech 1981 Kraft Drive, Blacksburg, VA 24061
- Fralin Life Sciences Institute, Virginia Tech 1981 Kraft Drive, Blacksburg, VA 24061
- Carilion School of Medicine, Virginia Tech 1981 Kraft Drive, Blacksburg, VA 24061
- Division of Systems Biology/AIS, Virginia Tech 1981 Kraft Drive, Blacksburg, VA 24061
| |
Collapse
|
9
|
Nielsen CM, Zhang X, Raygor K, Wang S, Bollen AW, Wang RA. Endothelial Rbpj deletion normalizes Notch4-induced brain arteriovenous malformation in mice. J Exp Med 2022; 220:213722. [PMID: 36441145 PMCID: PMC9700524 DOI: 10.1084/jem.20211390] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 10/10/2022] [Accepted: 11/09/2022] [Indexed: 11/29/2022] Open
Abstract
Upregulation of Notch signaling is associated with brain arteriovenous malformation (bAVM), a disease that lacks pharmacological treatments. Tetracycline (tet)-regulatable endothelial expression of constitutively active Notch4 (Notch4*tetEC) from birth induced bAVMs in 100% of mice by P16. To test whether targeting downstream signaling, while sustaining the causal Notch4*tetEC expression, induces AVM normalization, we deleted Rbpj, a mediator of Notch signaling, in endothelium from P16, by combining tet-repressible Notch4*tetEC with tamoxifen-inducible Rbpj deletion. Established pathologies, including AV connection diameter, AV shunting, vessel tortuosity, intracerebral hemorrhage, tissue hypoxia, life expectancy, and arterial marker expression were improved, compared with Notch4*tetEC mice without Rbpj deletion. Similarly, Rbpj deletion from P21 induced advanced bAVM regression. After complete AVM normalization induced by repression of Notch4*tetEC, virtually no bAVM relapsed, despite Notch4*tetEC re-expression in adults. Thus, inhibition of endothelial Rbpj halted Notch4*tetEC bAVM progression, normalized bAVM abnormalities, and restored microcirculation, providing proof of concept for targeting a downstream mediator to treat AVM pathologies despite a sustained causal molecular lesion.
Collapse
Affiliation(s)
- Corinne M. Nielsen
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Xuetao Zhang
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Kunal Raygor
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Shaoxun Wang
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Andrew W. Bollen
- Department of Pathology, University of California, San Francisco, San Francisco, CA
| | - Rong A. Wang
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California, San Francisco, San Francisco, CA,Correspondence to Rong A. Wang:
| |
Collapse
|
10
|
Li Y, Ma T, Zhu X, Zhang M, Zhao L, Wang P, Liang J. Zinc improves neurological recovery by promoting angiogenesis via the astrocyte-mediated HIF-1α/VEGF signaling pathway in experimental stroke. CNS Neurosci Ther 2022; 28:1790-1799. [PMID: 35855611 PMCID: PMC9532912 DOI: 10.1111/cns.13918] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Ischemic stroke is a serious cerebrovascular disease with high morbidity and disability. Zinc accumulation has been shown to play a vital role in neuronal death and blood-brain barrier damage following ischemia in acute stage. However, almost nothing is known about whether zinc is involved in neurological recovery in ischemic prolonged period. This study investigates whether zinc promotes neurological recovery through astrocytes-induced angiogenesis during ischemic repair phase. METHODS Sprague-Dawley rats were subjected to 2 h ischemia/14, 21, and 28 days reperfusion by middle cerebral artery occlusion, then administered ZnCl2 (10 mg/kg) via intraperitoneally daily from 7 days to tissue collection to observe brain tissue morphology, neurological function recovery by cortical width index, Adhesive removal test, and Forelimb placing test. Angiogenesis, astrocyte activation, and HIF-1α/VEGF pathway were assessed via Western blot, immunofluorescence, and BrdU method in vivo and in vitro. RESULTS The results showed that zinc significantly alleviated brain atrophy and improved neurological function recovery during the cerebral ischemia repair stage. Zinc significantly increased the protein levels of HIF-1α, VEGF-A, and VEGF-R2 in astrocytes, and promoted angiogenesis during cerebral ischemia repair. In vitro and in vivo studies confirmed that zinc promoted angiogenesis via the astrocyte-mediated HIF-1α/VEGF signaling pathway. CONCLUSIONS Zinc significantly improves neurological function recovery during the cerebral ischemia repair stage, providing new evidence supporting zinc as a potential therapeutic target for ischemic stroke by promoting astrocyte induced angiogenesis.
Collapse
Affiliation(s)
- Yang Li
- Institution of Life Science, Jinzhou Medical University, Jinzhou, China
| | - Tingting Ma
- Institution of Life Science, Jinzhou Medical University, Jinzhou, China
| | - Xiaoyu Zhu
- Institution of Life Science, Jinzhou Medical University, Jinzhou, China
| | - Mingqi Zhang
- Institution of Life Science, Jinzhou Medical University, Jinzhou, China
| | - Liang Zhao
- College of Pharmacy, Jinzhou Medical University, Jinzhou, China
| | - Peng Wang
- Key Laboratory of Neurodegenerative Diseases of Liaoning Province, Department of Neurobiology, Jinzhou Medical University, Jinzhou, China
| | - Jia Liang
- Institution of Life Science, Jinzhou Medical University, Jinzhou, China.,Key Laboratory of Neurodegenerative Diseases of Liaoning Province, Department of Neurobiology, Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
11
|
Alfieri A, Koudelka J, Li M, Scheffer S, Duncombe J, Caporali A, Kalaria RN, Smith C, Shah AM, Horsburgh K. Nox2 underpins microvascular inflammation and vascular contributions to cognitive decline. J Cereb Blood Flow Metab 2022; 42:1176-1191. [PMID: 35102790 PMCID: PMC9207496 DOI: 10.1177/0271678x221077766] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 11/26/2021] [Accepted: 12/06/2021] [Indexed: 11/16/2022]
Abstract
Chronic microvascular inflammation and oxidative stress are inter-related mechanisms underpinning white matter disease and vascular cognitive impairment (VCI). A proposed mediator is nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (Nox2), a major source of reactive oxygen species (ROS) in the brain. To assess the role of Nox2 in VCI, we studied a tractable model with white matter pathology and cognitive impairment induced by bilateral carotid artery stenosis (BCAS). Mice with genetic deletion of Nox2 (Nox2 KO) were compared to wild-type (WT) following BCAS. Sustained BCAS over 12 weeks in WT mice induced Nox2 expression, indices of microvascular inflammation and oxidative damage, along with white matter pathology culminating in a marked cognitive impairment, which were all protected by Nox2 genetic deletion. Neurovascular coupling was impaired in WT mice post-BCAS and restored in Nox2 KO mice. Increased vascular expression of chemoattractant mediators, cell-adhesion molecules and endothelial activation factors in WT mice post-BCAS were ameliorated by Nox2 deficiency. The clinical relevance was confirmed by increased vascular Nox2 and indices of microvascular inflammation in human post-mortem subjects with cerebral vascular disease. Our results support Nox2 activity as a critical determinant of VCI, whose targeting may be of therapeutic benefit in cerebral vascular disease.
Collapse
Affiliation(s)
- Alessio Alfieri
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- National Heart and Lung Institute, Vascular Science, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, UK
| | - Juraj Koudelka
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Mosi Li
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Sanny Scheffer
- Department of Pathology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Jessica Duncombe
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Andrea Caporali
- British Heart Foundation Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Rajesh N Kalaria
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle-Upon-Tyne, UK
| | - Colin Smith
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Ajay M Shah
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King’s College London, London, UK
| | - Karen Horsburgh
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
12
|
Chapman AD, Selhorst S, LaComb J, LeDantec-Boswell A, Wohl TR, Adhicary S, Nielsen CM. Endothelial Rbpj Is Required for Cerebellar Morphogenesis and Motor Control in the Early Postnatal Mouse Brain. CEREBELLUM (LONDON, ENGLAND) 2022:10.1007/s12311-022-01429-w. [PMID: 35716334 DOI: 10.1007/s12311-022-01429-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/07/2022] [Indexed: 11/25/2022]
Abstract
Intercellular influences are necessary for coordinated development and function of vascular and neural components in the brain. In the early postnatal period after birth, the mammalian cerebellum undergoes extensive morphogenesis - developing its characteristic lobules, organizing its diverse cell types into defined cellular layers, and establishing neural circuits that support cerebellar function, such as coordinated movement. In parallel, the cerebellar vasculature undergoes extensive postnatal growth and maturation, keeping pace with the expanding neural compartment. Endothelial deletion of Rbpj leads to neurovascular abnormalities in mice, including arteriovenous (AV) shunts that supplant capillaries and instead direct high-pressure/high-flow arterial blood directly to veins. Gross and histopathological cerebellar abnormalities, associated with these Rbpj-mediated brain AV malformations (AVMs), led to our hypothesis that early postnatal morphogenesis and lamination of cerebellum was perturbed in mice harboring endothelial Rbpj deficiency from birth. Here, we show that endothelial Rbpj-mutant mice developed enlarged vascular malformations on the cerebellar surface, by 2-week post-Rbpj deletion. In addition, outgrowth of cerebellar lobules was impaired through decreased cell proliferation, but not increased apoptosis, in the external granule layer. Molecular layer thickness was reduced, and the Purkinje layer was affected, by decreased Purkinje cell number, primary dendrite length, and dendritic arbor density. Endothelial deletion of Rbpj also led to impaired motor behaviors, consistent with abnormal cerebellar morphogenesis and lamination. Thus, our data suggest that Rbpj is required, in early postnatal vascular endothelium, to ensure proper cerebellar outgrowth, morphogenesis, and function in mice.
Collapse
Affiliation(s)
- Amelia D Chapman
- Department of Biological Sciences, Ohio University, 57 Oxbow Trail, Irvine Hall 107, Athens, OH, 45701, USA
- Honors Tutorial College, Ohio University, Athens, OH, 45701, USA
| | - Samantha Selhorst
- Department of Biological Sciences, Ohio University, 57 Oxbow Trail, Irvine Hall 107, Athens, OH, 45701, USA
- Honors Tutorial College, Ohio University, Athens, OH, 45701, USA
| | - Julia LaComb
- Department of Biological Sciences, Ohio University, 57 Oxbow Trail, Irvine Hall 107, Athens, OH, 45701, USA
| | - Alexis LeDantec-Boswell
- Department of Biological Sciences, Ohio University, 57 Oxbow Trail, Irvine Hall 107, Athens, OH, 45701, USA
| | - Timothy R Wohl
- Department of Biological Sciences, Ohio University, 57 Oxbow Trail, Irvine Hall 107, Athens, OH, 45701, USA
- Honors Tutorial College, Ohio University, Athens, OH, 45701, USA
| | - Subhodip Adhicary
- Department of Biological Sciences, Ohio University, 57 Oxbow Trail, Irvine Hall 107, Athens, OH, 45701, USA
- Translational Biomedical Sciences Program, Ohio University, Athens, OH, 45701, USA
| | - Corinne M Nielsen
- Department of Biological Sciences, Ohio University, 57 Oxbow Trail, Irvine Hall 107, Athens, OH, 45701, USA.
- Molecular and Cellular Biology Program, Ohio University, Athens, OH, 45701, USA.
- Neuroscience Program, Ohio University, Athens, OH, 45701, USA.
| |
Collapse
|
13
|
Puebla M, Tapia PJ, Espinoza H. Key Role of Astrocytes in Postnatal Brain and Retinal Angiogenesis. Int J Mol Sci 2022; 23:ijms23052646. [PMID: 35269788 PMCID: PMC8910249 DOI: 10.3390/ijms23052646] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 01/27/2023] Open
Abstract
Angiogenesis is a key process in various physiological and pathological conditions in the nervous system and in the retina during postnatal life. Although an increasing number of studies have addressed the role of endothelial cells in this event, the astrocytes contribution in angiogenesis has received less attention. This review is focused on the role of astrocytes as a scaffold and in the stabilization of the new blood vessels, through different molecules release, which can modulate the angiogenesis process in the brain and in the retina. Further, differences in the astrocytes phenotype are addressed in glioblastoma, one of the most devastating types of brain cancer, in order to provide potential targets involved in the cross signaling between endothelial cells, astrocytes and glioma cells, that mediate tumor progression and pathological angiogenesis. Given the relevance of astrocytes in angiogenesis in physiological and pathological conditions, future studies are required to better understand the interrelation between endothelial and astrocyte signaling pathways during this process.
Collapse
Affiliation(s)
- Mariela Puebla
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina-Clínica Alemana, Universidad del Desarrollo, Av. Plaza 680, Las Condes, Santiago 7550000, Chile;
| | - Pablo J. Tapia
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Av. Lota 2465, Providencia, Santiago 7500000, Chile;
- Facultad de Medicina Veterinaria y Agronomía, Universidad de las Américas, Av. República 71, Santiago 8320000, Chile
| | - Hilda Espinoza
- Facultad de Ciencias de la Salud, Universidad del Alba, Av. Ejército Libertador 171, Santiago 8320000, Chile
- Correspondence:
| |
Collapse
|
14
|
Martončíková M, Alexovič Matiašová A, Ševc J, Račeková E. Relationship between Blood Vessels and Migration of Neuroblasts in the Olfactory Neurogenic Region of the Rodent Brain. Int J Mol Sci 2021; 22:11506. [PMID: 34768936 PMCID: PMC8583928 DOI: 10.3390/ijms222111506] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 11/25/2022] Open
Abstract
Neural precursors originating in the subventricular zone (SVZ), the largest neurogenic region of the adult brain, migrate several millimeters along a restricted migratory pathway, the rostral migratory stream (RMS), toward the olfactory bulb (OB), where they differentiate into interneurons and integrate into the local neuronal circuits. Migration of SVZ-derived neuroblasts in the adult brain differs in many aspects from that in the embryonic period. Unlike in that period, postnatally-generated neuroblasts in the SVZ are able to divide during migration along the RMS, as well as they migrate independently of radial glia. The homophilic mode of migration, i.e., using each other to move, is typical for neuroblast movement in the RMS. In addition, it has recently been demonstrated that specifically-arranged blood vessels navigate SVZ-derived neuroblasts to the OB and provide signals which promote migration. Here we review the development of vasculature in the presumptive neurogenic region of the rodent brain during the embryonic period as well as the development of the vascular scaffold guiding neuroblast migration in the postnatal period, and the significance of blood vessel reorganization during the early postnatal period for proper migration of RMS neuroblasts in adulthood.
Collapse
Affiliation(s)
- Marcela Martončíková
- Department of Regenerative Medicine and Cell Therapy, Institute of Neurobiology, Biomedical Research Center, Slovak Academy of Sciences, Šoltésovej 4, 040 01 Košice, Slovakia;
| | - Anna Alexovič Matiašová
- Department of Cell Biology, Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafárik University in Košice, Šrobárova 2, 041 54 Košice, Slovakia; (A.A.M.); (J.Š.)
| | - Juraj Ševc
- Department of Cell Biology, Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafárik University in Košice, Šrobárova 2, 041 54 Košice, Slovakia; (A.A.M.); (J.Š.)
| | - Enikő Račeková
- Department of Regenerative Medicine and Cell Therapy, Institute of Neurobiology, Biomedical Research Center, Slovak Academy of Sciences, Šoltésovej 4, 040 01 Košice, Slovakia;
| |
Collapse
|
15
|
Lee S, Lee HS, Chung JJ, Kim SH, Park JW, Lee K, Jung Y. Enhanced Regeneration of Vascularized Adipose Tissue with Dual 3D-Printed Elastic Polymer/dECM Hydrogel Complex. Int J Mol Sci 2021; 22:ijms22062886. [PMID: 33809175 PMCID: PMC7999751 DOI: 10.3390/ijms22062886] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 12/11/2022] Open
Abstract
A flexible and bioactive scaffold for adipose tissue engineering was fabricated and evaluated by dual nozzle three-dimensional printing. A highly elastic poly (L-lactide-co-ε-caprolactone) (PLCL) copolymer, which acted as the main scaffolding, and human adipose tissue derived decellularized extracellular matrix (dECM) hydrogels were used as the printing inks to form the scaffolds. To prepare the three-dimensional (3D) scaffolds, the PLCL co-polymer was printed with a hot melting extruder system while retaining its physical character, similar to adipose tissue, which is beneficial for regeneration. Moreover, to promote adipogenic differentiation and angiogenesis, adipose tissue-derived dECM was used. To optimize the printability of the hydrogel inks, a mixture of collagen type I and dECM hydrogels was used. Furthermore, we examined the adipose tissue formation and angiogenesis of the PLCL/dECM complex scaffold. From in vivo experiments, it was observed that the matured adipose-like tissue structures were abundant, and the number of matured capillaries was remarkably higher in the hydrogel–PLCL group than in the PLCL-only group. Moreover, a higher expression of M2 macrophages, which are known to be involved in the remodeling and regeneration of tissues, was detected in the hydrogel–PLCL group by immunofluorescence analysis. Based on these results, we suggest that our PLCL/dECM fabricated by a dual 3D printing system will be useful for the treatment of large volume fat tissue regeneration.
Collapse
Affiliation(s)
- Soojin Lee
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Korea; (S.L.); (J.J.C.); (S.H.K.)
- Program in Nanoscience and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Korea;
| | - Hyun Su Lee
- Program in Nanoscience and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Korea;
| | - Justin J. Chung
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Korea; (S.L.); (J.J.C.); (S.H.K.)
| | - Soo Hyun Kim
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Korea; (S.L.); (J.J.C.); (S.H.K.)
- NBIT, KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| | - Jong Woong Park
- Department of Orthopedic Surgery, Korea University Anam Hospital, Seoul 02841, Korea;
| | - Kangwon Lee
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Korea
- Correspondence: (K.L.); (Y.J.)
| | - Youngmee Jung
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Korea; (S.L.); (J.J.C.); (S.H.K.)
- School of Electrical and Electronic Engineering, YU-KIST Institute, Yonsei University, Seoul 03722, Korea
- Correspondence: (K.L.); (Y.J.)
| |
Collapse
|
16
|
Winkelman MA, Koppes AN, Koppes RA, Dai G. Bioengineering the neurovascular niche to study the interaction of neural stem cells and endothelial cells. APL Bioeng 2021; 5:011507. [PMID: 33688617 PMCID: PMC7932757 DOI: 10.1063/5.0027211] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 02/15/2021] [Indexed: 12/13/2022] Open
Abstract
The ability of mammalian neural stem cells (NSCs) to self-renew and differentiate throughout adulthood has made them ideal to study neurogenesis and attractive candidates for neurodegenerative disease therapies. In the adult mammalian brain, NSCs are maintained in the neurovascular niche (NVN) where they are found near the specialized blood vessels, suggesting that brain endothelial cells (BECs) are prominent orchestrators of NSC fate. However, most of the current knowledge of the mammalian NVN has been deduced from nonhuman studies. To circumvent the challenges of in vivo studies, in vitro models have been developed to better understand the reciprocal cellular mechanisms of human NSCs and BECs. This review will cover the current understanding of mammalian NVN biology, the effects of endothelial cell-derived signals on NSC fate, and the in vitro models developed to study the interactions between NSCs and BECs.
Collapse
Affiliation(s)
- Max A Winkelman
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, USA
| | | | - Ryan A Koppes
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, USA
| | - Guohao Dai
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, USA
| |
Collapse
|
17
|
Du Preez A, Onorato D, Eiben I, Musaelyan K, Egeland M, Zunszain PA, Fernandes C, Thuret S, Pariante CM. Chronic stress followed by social isolation promotes depressive-like behaviour, alters microglial and astrocyte biology and reduces hippocampal neurogenesis in male mice. Brain Behav Immun 2021; 91:24-47. [PMID: 32755644 DOI: 10.1016/j.bbi.2020.07.015] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 06/29/2020] [Accepted: 07/11/2020] [Indexed: 12/11/2022] Open
Abstract
Unpredictable chronic mild stress (UCMS) is one of the most commonly used, robust and translatable models for studying the neurobiological basis of major depression. Although the model currently has multiple advantages, it does not entirely follow the trajectory of the disorder, whereby depressive symptomology can often present months after exposure to stress. Furthermore, patients with depression are more likely to withdraw in response to their stressful experience, or as a symptom of their depression, and, in turn, this withdrawal/isolation can further exacerbate the stressful experience and the depressive symptomology. Therefore, we investigated the effect(s) of 6 weeks of UCMS followed by another 6 weeks of social isolation (referred to as UCMSI), on behaviour, corticosterone stress responsivity, immune system functioning, and hippocampal neurogenesis, in young adult male mice. We found that UCMSI induced several behavioural changes resembling depression but did not induce peripheral inflammation. However, UCMSI animals showed increased microglial activation in the ventral dentate gyrus (DG) of the hippocampus and astrocyte activation in both the dorsal and ventral DG, with increased GFAP-positive cell immunoreactivity, GFAP-positive cell hypertrophy and process extension, and increased s100β-positive cell density. Moreover, UCMSI animals had significantly reduced neurogenesis in the DG and reduced levels of peripheral vascular endothelial growth factor (VEGF) - a trophic factor produced by astrocytes and that stimulates neurogenesis. Finally, UCMSI mice also had normal baseline corticosterone levels but a smaller increase in corticosterone following acute stress, that is, the Porsolt Swim Test. Our work gives clinically relevant insights into the role that microglial and astrocyte functioning, and hippocampal neurogenesis may play in the context of stress, social isolation and depression, offering a potentially new avenue for therapeutic target.
Collapse
Affiliation(s)
- Andrea Du Preez
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK.
| | - Diletta Onorato
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Inez Eiben
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Ksenia Musaelyan
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Martin Egeland
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Patricia A Zunszain
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Cathy Fernandes
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK; MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Sandrine Thuret
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Carmine M Pariante
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| |
Collapse
|
18
|
Du Preez A, Law T, Onorato D, Lim YM, Eiben P, Musaelyan K, Egeland M, Hye A, Zunszain PA, Thuret S, Pariante CM, Fernandes C. The type of stress matters: repeated injection and permanent social isolation stress in male mice have a differential effect on anxiety- and depressive-like behaviours, and associated biological alterations. Transl Psychiatry 2020; 10:325. [PMID: 32958745 PMCID: PMC7505042 DOI: 10.1038/s41398-020-01000-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/12/2020] [Accepted: 09/03/2020] [Indexed: 01/02/2023] Open
Abstract
Chronic stress can alter the immune system, adult hippocampal neurogenesis and induce anxiety- and depressive-like behaviour in rodents. However, previous studies have not discriminated between the effect(s) of different types of stress on these behavioural and biological outcomes. We investigated the effect(s) of repeated injection vs. permanent social isolation on behaviour, stress responsivity, immune system functioning and hippocampal neurogenesis, in young adult male mice, and found that the type of stress exposure does indeed matter. Exposure to 6 weeks of repeated injection resulted in an anxiety-like phenotype, decreased systemic inflammation (i.e., reduced plasma levels of TNFα and IL4), increased corticosterone reactivity, increased microglial activation and decreased neuronal differentiation in the dentate gyrus (DG). In contrast, exposure to 6 weeks of permanent social isolation resulted in a depressive-like phenotype, increased plasma levels of TNFα, decreased plasma levels of IL10 and VEGF, decreased corticosterone reactivity, decreased microglial cell density and increased cell density for radial glia, s100β-positive cells and mature neuroblasts-all in the DG. Interestingly, combining the two distinct stress paradigms did not have an additive effect on behavioural and biological outcomes, but resulted in yet a different phenotype, characterized by increased anxiety-like behaviour, decreased plasma levels of IL1β, IL4 and VEGF, and decreased hippocampal neuronal differentiation, without altered neuroinflammation or corticosterone reactivity. These findings demonstrate that different forms of chronic stress can differentially alter both behavioural and biological outcomes in young adult male mice, and that combining multiple stressors may not necessarily cause more severe pathological outcomes.
Collapse
Affiliation(s)
- Andrea Du Preez
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Thomas Law
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Diletta Onorato
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Yau M Lim
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Paola Eiben
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Ksenia Musaelyan
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Martin Egeland
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Abdul Hye
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Patricia A Zunszain
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Sandrine Thuret
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Carmine M Pariante
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Cathy Fernandes
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
| |
Collapse
|
19
|
Detter MR, Shenkar R, Benavides CR, Neilson CA, Moore T, Lightle R, Hobson N, Shen L, Cao Y, Girard R, Zhang D, Griffin E, Gallione CJ, Awad IA, Marchuk DA. Novel Murine Models of Cerebral Cavernous Malformations. Angiogenesis 2020; 23:651-666. [PMID: 32710309 DOI: 10.1007/s10456-020-09736-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/06/2020] [Indexed: 12/21/2022]
Abstract
Cerebral cavernous malformations (CCMs) are ectatic capillary-venous malformations that develop in approximately 0.5% of the population. Patients with CCMs may develop headaches, focal neurologic deficits, seizures, and hemorrhages. While symptomatic CCMs, depending upon the anatomic location, can be surgically removed, there is currently no pharmaceutical therapy to treat CCMs. Several mouse models have been developed to better understand CCM pathogenesis and test therapeutics. The most common mouse models induce a large CCM burden that is anatomically restricted to the cerebellum and contributes to lethality in the early days of life. These inducible models thus have a relatively short period for drug administration. We developed an inducible CCM3 mouse model that develops CCMs after weaning and provides a longer period for potential therapeutic intervention. Using this new model, three recently proposed CCM therapies, fasudil, tempol, vitamin D3, and a combination of the three drugs, failed to substantially reduce CCM formation when treatment was administered for 5 weeks, from postnatal day 21 (P21) to P56. We next restricted Ccm3 deletion to the brain vasculature and provided greater time (121 days) for CCMs to develop chronic hemorrhage, recapitulating the human lesions. We also developed the first model of acute CCM hemorrhage by injecting mice harboring CCMs with lipopolysaccharide. These efficient models will enable future drug studies to more precisely target clinically relevant features of CCM disease: CCM formation, chronic hemorrhage, and acute hemorrhage.
Collapse
Affiliation(s)
- Matthew R Detter
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27705, USA
| | - Robert Shenkar
- Neurovascular Surgery Program, Department of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, 60637, USA
| | - Christian R Benavides
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27705, USA
| | - Catherine A Neilson
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27705, USA
| | - Thomas Moore
- Neurovascular Surgery Program, Department of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, 60637, USA
| | - Rhonda Lightle
- Neurovascular Surgery Program, Department of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, 60637, USA
| | - Nicholas Hobson
- Neurovascular Surgery Program, Department of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, 60637, USA
| | - Le Shen
- Neurovascular Surgery Program, Department of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, 60637, USA
| | - Ying Cao
- Neurovascular Surgery Program, Department of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, 60637, USA
| | - Romuald Girard
- Neurovascular Surgery Program, Department of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, 60637, USA
| | - Dongdong Zhang
- Neurovascular Surgery Program, Department of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, 60637, USA
| | - Erin Griffin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27705, USA
| | - Carol J Gallione
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27705, USA
| | - Issam A Awad
- Neurovascular Surgery Program, Department of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, 60637, USA
| | - Douglas A Marchuk
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27705, USA. .,James B Duke Professor, Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Box 3175, Durham, NC, 27710, USA.
| |
Collapse
|
20
|
Lee LKC, Leong LI, Liu Y, Luo M, Chan HYE, Choi CHJ. Preclinical Nanomedicines for Polyglutamine-Based Neurodegenerative Diseases. Mol Pharm 2020; 18:610-626. [PMID: 32584043 DOI: 10.1021/acs.molpharmaceut.0c00506] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Polyglutamine (polyQ) diseases, such as Huntington's disease and several types of spinocerebellar ataxias, are dominantly inherited progressive neurodegenerative disorders and characterized by the presence of expanded CAG trinucleotide repeats in the respective disease locus of the patient genomes. Patients with polyQ diseases currently need to rely on symptom-relieving treatments because disease-modifying therapeutic interventions remain scarce. Many disease-modifying therapeutic agents are now under clinical testing for treating polyQ diseases, but their delivery to the brain is often too invasive (e.g., intracranial injection) or inefficient, owing to in vivo degradation and clearance by physiological barriers (e.g., oral and intravenous administration). Nanoparticles provide a feasible solution for improving drug delivery to the brain, as evidenced by an increasing number of preclinical studies that document the efficacy of nanomedicines for polyQ diseases over the past 5-6 years. In this review, we present the pathogenic mechanisms of polyQ diseases, the common animal models of polyQ diseases for evaluating the efficacy of nanomedicines, and the common administration routes for delivering nanoparticles to the brain. Next, we summarize the recent preclinical applications of nanomedicines for treating polyQ diseases and improving neurological conditions in vivo, placing emphasis on antisense oligonucleotides, small peptide inhibitors, and small molecules as the disease-modifying agents. We conclude with our perspectives of the burgeoning field of "nanomedicines for polyQ diseases", including the use of inorganic nanoparticles and potential drugs as next-generation nanomedicines, development of higher-order animal models of polyQ diseases, and importance of "brain-nano" interactions.
Collapse
Affiliation(s)
| | | | | | - Meihua Luo
- Monash Institute of Pharmaceutics Science, Monash University, Parkville, Victoria 3052, Australia
| | | | | |
Collapse
|
21
|
Ardalan M, Elfving B, Rafati AH, Mansouri M, Zarate CA, Mathe AA, Wegener G. Rapid effects of S-ketamine on the morphology of hippocampal astrocytes and BDNF serum levels in a sex-dependent manner. Eur Neuropsychopharmacol 2020; 32:94-103. [PMID: 31973999 PMCID: PMC7281850 DOI: 10.1016/j.euroneuro.2020.01.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 12/13/2019] [Accepted: 01/02/2020] [Indexed: 12/16/2022]
Abstract
The prevalence of major depressive disorder (MDD) is higher in women than men. Importantly, a differential behavioral response by sex to the antidepressant response to ketamine in rodents has been reported. Mechanistically, male depressed-like animals showed an increased spine density after ketamine treatment via restoration of synaptic protein levels while those proteins were not altered in female rats. In addition, preclinical studies indicate that the impairment of astrocytic plasticity is one of the contributing mechanisms in the pathophysiology of MDD. Accordingly, in this study, we determined the effect of sex on the rapid morphological alteration of hippocampal astrocytes and the serum level of BDNF one hour after S-ketamine injection. A single intraperitoneal dose of S-ketamine (15 mg/kg) or saline was injected to the male and female Flinders Sensitive Line (FSL) rats, a genetic animal model of depression and their brains were perfused one hour after treatment. The size of the GFAP positive astrocytes in the hippocampal subregions was measured. The volume of different hippocampal subregions was assessed using the Cavalieri estimator. Moreover, serum levels of BDNF were measured with enzyme-linked immunosorbent assay (ELISA) kits. The volume of hippocampal subregions significantly increased one hour after S-ketamine in both male and female FSL animals. However, a substantial alteration in the morphology of the hippocampal astrocytes was observed only in the female rats. Additionally, significantly increased serum BDNF levels in the female depressed rats were observed one hour after S-ketamine treatment. Our results indicate that the rapid effects of S-ketamine on the morphology of the hippocampal astrocytes and the serum level of BDNF are sex-dependent.
Collapse
Affiliation(s)
- Maryam Ardalan
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Clinical Medicine, Center of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark.
| | - Betina Elfving
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Ali H Rafati
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Monireh Mansouri
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Institute for Cognitive and Brain Sciences, Shahid Beheshti University, Tehran, Iran
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National, Institute of Mental Health, National Institutes of Health, Bethesda, USA
| | - Aleksander A Mathe
- Department of Clinical Neuroscience, Karolinska Institutet, Stockohlm, Sweden
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Center of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa; AUGUST Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
22
|
Mayegowda SB, Thomas C. Glial pathology in neuropsychiatric disorders: a brief review. J Basic Clin Physiol Pharmacol 2019; 30:jbcpp-2018-0120. [PMID: 31280243 DOI: 10.1515/jbcpp-2018-0120] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 01/24/2019] [Indexed: 06/09/2023]
Abstract
Neurons have been considered the major functional entities of the nervous system that are responsible for most of the functions even though glial cells largely outnumber them. However, recent reports have proved that glial cells do not function just like glue in the nervous system but also substantially affect neuronal function and activities, and are significantly involved in the underlying pathobiology of various psychiatric disorders. Dysfunctional astrocytes and degeneration of glial cells are postulated to be critical factors contributing to the aggravation of depressive-like symptoms in humans, which was proved using animal models. Alteration in glial cell function predominantly targets three main brain regions - the prefrontal cortex, limbic areas including the hippocampus, and the amygdala, which have been extensively studied by various researchers across the globe. These studies have postulated that failure in adopting to the changing neurophysiology due to stress will lead to regressive plasticity in the hippocampus and prefrontal cortex, but to progressive plasticity in the amygdala. In this present review, an effort has been made to understand the different alterations in chronic stress models in correlation with clinical conditions, providing evidence on the defective maintenance of glial function and its potential role in the precipitation of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Shilpa Borehalli Mayegowda
- School of Basic and Applied Sciences, Dayananda Sagar University, Shavige Malleshwara Hills, Kumaraswamy Layout,Bengaluru 560 078, India, Mobile: +919972939466
| | - Christofer Thomas
- Department of Physiology, Sapthagiri Institute of Medical Sciences and Research Centre, Bengaluru, India
| |
Collapse
|
23
|
Koester-Hegmann C, Bengoetxea H, Kosenkov D, Thiersch M, Haider T, Gassmann M, Schneider Gasser EM. High-Altitude Cognitive Impairment Is Prevented by Enriched Environment Including Exercise via VEGF Signaling. Front Cell Neurosci 2019; 12:532. [PMID: 30687018 PMCID: PMC6335396 DOI: 10.3389/fncel.2018.00532] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 12/24/2018] [Indexed: 01/17/2023] Open
Abstract
Exposure to hypobaric hypoxia at high altitude (above 2500 m asl) causes cognitive impairment, mostly attributed to changes in brain perfusion and consequently neuronal death. Enriched environment and voluntary exercise has been shown to improve cognitive function, to enhance brain microvasculature and neurogenesis, and to be neuroprotective. Here we show that high-altitude exposure (3540 m asl) of Long Evans rats during early adulthood (P48-P59) increases brain microvasculature and neurogenesis but impairs spatial and visual memory along with an increase in neuronal apoptosis. We tested whether enriched environment including a running wheel for voluntary exercise (EE) can prevent cognitive impairment at high-altitude and whether apoptosis is prevented. We found that EE retained spatial and visual memory at high altitude, and prevented neuronal apoptosis. Further, we tested whether vascular endothelial growth factor (VEGF) signaling is required for the EE-mediated recovery of spatial and visual memory and the reduction in apoptosis. Pharmacological inhibition of VEGF signaling by oral application of a tyrosine kinase inhibitor (Vandetanib) prevented the recovery of spatial and visual memory in animals housed in EE, along with an increase in apoptosis and a reduction in neurogenesis. Surprisingly, inhibition of VEGF signaling also caused impairment in spatial memory in EE-housed animals reared at low altitude, affecting mainly dentate gyrus microvasculature but not neurogenesis. We conclude that EE-mediated VEGF signaling is neuroprotective and essential for the maintenance of cognition and neurogenesis during high-altitude exposure, and for the maintenance of spatial memory at low altitude. Finally, our data also underlines the potential risk of cognitive impairment and disturbed high altitude adaption from the use of VEGF-signaling inhibitors for therapeutic purposes.
Collapse
Affiliation(s)
- Christina Koester-Hegmann
- Neuroprotection Group, Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Harkaitz Bengoetxea
- Laboratory of Clinical and Experimental Neuroscience (LaNCE), Department of Neuroscience, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Dmitry Kosenkov
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Markus Thiersch
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Thomas Haider
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,Universidad Peruana Cayetano Heredia (UPCH), Lima, Peru
| | - Edith M Schneider Gasser
- Neuroprotection Group, Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland.,Center for Neuroscience Zurich (ZNZ), Zurich, Switzerland
| |
Collapse
|
24
|
Wang F, Cao Y, Ma L, Pei H, Rausch WD, Li H. Dysfunction of Cerebrovascular Endothelial Cells: Prelude to Vascular Dementia. Front Aging Neurosci 2018; 10:376. [PMID: 30505270 PMCID: PMC6250852 DOI: 10.3389/fnagi.2018.00376] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/29/2018] [Indexed: 12/19/2022] Open
Abstract
Vascular dementia (VaD) is the second most common type of dementia after Alzheimer's disease (AD), characterized by progressive cognitive impairment, memory loss, and thinking or speech problems. VaD is usually caused by cerebrovascular disease, during which, cerebrovascular endothelial cells (CECs) are vulnerable. CEC dysfunction occurs before the onset of VaD and can eventually lead to dysregulation of cerebral blood flow and blood-brain barrier damage, followed by the activation of glia and inflammatory environment in the brain. White matter, neuronal axons, and synapses are compromised in this process, leading to cognitive impairment. The present review summarizes the mechanisms underlying CEC impairment during hypoperfusion and pathological role of CECs in VaD. Through the comprehensive examination and summarization, endothelial nitric oxide synthase (eNOS)/nitric oxide (NO) signaling pathway, Ras homolog gene family member A (RhoA) signaling pathway, and CEC-derived caveolin-1 (CAV-1) are proposed to serve as targets of new drugs for the treatment of VaD.
Collapse
Affiliation(s)
- Feixue Wang
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Cao
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Lina Ma
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Hui Pei
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Wolf Dieter Rausch
- Department for Biomedical Sciences, Institute of Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Hao Li
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
25
|
Pérez Ortiz JM, Orr HT. Spinocerebellar Ataxia Type 1: Molecular Mechanisms of Neurodegeneration and Preclinical Studies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1049:135-145. [PMID: 29427101 DOI: 10.1007/978-3-319-71779-1_6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Spinocerebellar ataxia type 1 (SCA1) is an adult-onset, inherited disease that leads to degeneration of Purkinje cells of the cerebellum and culminates in death 10-30 years after disease onset. SCA1 is caused by a CAG repeat mutation in the ATXN1 gene, encoding the ATXN1 protein with an abnormally expanded polyglutamine tract. As neurodegeneration progresses, other brain regions become involved and contribute to cognitive deficits as well as problems with speech, swallowing, and control of breathing. The fundamental basis of pathology is an aberration in the normal function of Purkinje cells affecting regulation of gene transcription and RNA splicing. Glutamine-expanded ATXN1 is highly stable and more resistant to degradation. Moreover, phosphorylation at S776 in ATXN1 is a post-translational modification known to influence protein levels. SCA1 remains an untreatable disease managed only by palliative care. Preclinical studies are founded on the principle that mutant protein load is toxic and attenuating ATXN1 protein levels can alleviate disease. Two approaches being pursued are targeting gene expression or protein levels. Viral delivery of miRNAs harnesses the RNAi pathway to destroy ATXN1 mRNA. This approach shows promise in mouse models of disease. At the protein level, kinase inhibitors that block ATXN1-S776 phosphorylation may lead to therapeutic clearance of unphosphorylated ATXN1.
Collapse
Affiliation(s)
- Judit M Pérez Ortiz
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA.,Medical Scientist Training Program, University of Minnesota, Minneapolis, MN, 55455, USA.,Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Harry T Orr
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA. .,Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
26
|
Zhang N, Chen J, Ferraro GB, Wu L, Datta M, Jain RK, Plotkin SR, Stemmer-Rachamimov A, Xu L. Anti-VEGF treatment improves neurological function in tumors of the nervous system. Exp Neurol 2017; 299:326-333. [PMID: 28911884 DOI: 10.1016/j.expneurol.2017.09.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 07/05/2017] [Accepted: 09/10/2017] [Indexed: 01/17/2023]
Abstract
Research of various diseases of the nervous system has shown that VEGF has direct neuroprotective effects in the central and peripheral nervous systems, and indirect effects on improving neuronal vessel perfusion which leads to nerve protection. In the tumors of the nervous system, VEGF plays a critical role in tumor angiogenesis and tumor progression. The effect of anti-VEGF treatment on nerve protection and function has been recently reported - by normalizing the tumor vasculature, anti-VEGF treatment is able to relieve nerve edema and deliver oxygen more efficiently into the nerve, thus reducing nerve damage and improving nerve function. This review aims to summarize the divergent roles of VEGF in diseases of the nervous system and the recent findings of anti-VEGF therapy in nerve damage/regeneration and function in tumors, specifically, in Neurofibromatosis type 2 associated schwannomas.
Collapse
Affiliation(s)
- Na Zhang
- Edwin Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jie Chen
- Edwin Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Gino B Ferraro
- Edwin Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Limeng Wu
- Edwin Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Meenal Datta
- Edwin Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Department of Chemical and Biological Engineering, Tufts University, Medford, MA 02155, USA
| | - Rakesh K Jain
- Edwin Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Scott R Plotkin
- Department of Neurology and Cancer Center, Massachusetts General Hospital, USA
| | - Anat Stemmer-Rachamimov
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Lei Xu
- Edwin Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
27
|
Dezonne RS, Sartore RC, Nascimento JM, Saia-Cereda VM, Romão LF, Alves-Leon SV, de Souza JM, Martins-de-Souza D, Rehen SK, Gomes FCA. Derivation of Functional Human Astrocytes from Cerebral Organoids. Sci Rep 2017; 7:45091. [PMID: 28345587 PMCID: PMC5366860 DOI: 10.1038/srep45091] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 02/15/2017] [Indexed: 12/17/2022] Open
Abstract
Astrocytes play a critical role in the development and homeostasis of the central nervous system (CNS). Astrocyte dysfunction results in several neurological and degenerative diseases. However, a major challenge to our understanding of astrocyte physiology and pathology is the restriction of studies to animal models, human post-mortem brain tissues, or samples obtained from invasive surgical procedures. Here, we report a protocol to generate human functional astrocytes from cerebral organoids derived from human pluripotent stem cells. The cellular isolation of cerebral organoids yielded cells that were morphologically and functionally like astrocytes. Immunolabelling and proteomic assays revealed that human organoid-derived astrocytes express the main astrocytic molecular markers, including glutamate transporters, specific enzymes and cytoskeletal proteins. We found that organoid-derived astrocytes strongly supported neuronal survival and neurite outgrowth and responded to ATP through transient calcium wave elevations, which are hallmarks of astrocyte physiology. Additionally, these astrocytes presented similar functional pathways to those isolated from adult human cortex by surgical procedures. This is the first study to provide proteomic and functional analyses of astrocytes isolated from human cerebral organoids. The isolation of these astrocytes holds great potential for the investigation of developmental and evolutionary features of the human brain and provides a useful approach to drug screening and neurodegenerative disease modelling.
Collapse
Affiliation(s)
- Rômulo Sperduto Dezonne
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ., Brasil
| | - Rafaela Costa Sartore
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ., Brasil.,Instituto D'Or de Pesquisa e Ensino (IDOR), Rio de Janeiro, RJ, Brasil
| | - Juliana Minardi Nascimento
- Instituto D'Or de Pesquisa e Ensino (IDOR), Rio de Janeiro, RJ, Brasil.,Instituto de Biologia, Universidade Estadual de Campinas, Campinas, SP, Brasil
| | | | - Luciana Ferreira Romão
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ., Brasil.,Universidade Federal do Rio de Janeiro,Campus Xerém, RJ, Brasil
| | - Soniza Vieira Alves-Leon
- Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Jorge Marcondes de Souza
- Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | | | - Stevens Kastrup Rehen
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ., Brasil.,Instituto D'Or de Pesquisa e Ensino (IDOR), Rio de Janeiro, RJ, Brasil
| | | |
Collapse
|
28
|
Azizoglu DB, Chong DC, Villasenor A, Magenheim J, Barry DM, Lee S, Marty-Santos L, Fu S, Dor Y, Cleaver O. Vascular development in the vertebrate pancreas. Dev Biol 2016; 420:67-78. [PMID: 27789228 DOI: 10.1016/j.ydbio.2016.10.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 08/30/2016] [Accepted: 10/12/2016] [Indexed: 01/10/2023]
Abstract
The vertebrate pancreas is comprised of a highly branched tubular epithelium, which is intimately associated with an extensive and specialized vasculature. While we know a great deal about basic vascular anatomy of the adult pancreas, as well as islet capillaries, surprisingly little is known about the ontogeny of its blood vessels. Here, we analyze development of the pancreatic vasculature in the mouse embryo. We show that pancreatic epithelial branches intercalate with the fine capillary plexus of the surrounding pancreatic mesenchyme. Endothelial cells (ECs) within this mesenchyme are heterogeneous from the onset of organogenesis. Pancreatic arteries take shape before veins, in a manner analogous to early embryonic vessels. The main central artery forms during mid-gestation, as a result of vessel coalescence and remodeling of a vascular plexus. In addition, we show that vessels in the forming pancreas display a predictable architecture that is dependent on VEGF signaling. Over-expression of VEGF disrupts vascular patterning and arteriovenous differentiation within the developing pancreas. This study constitutes a first-time in-depth cellular and molecular characterization of pancreatic blood vessels, as they coordinately grow along with the pancreatic epithelium.
Collapse
Affiliation(s)
- D Berfin Azizoglu
- Department of Molecular Biology and Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Diana C Chong
- Department of Biology, University of North Carolina, Chapel Hill, NC, USA
| | - Alethia Villasenor
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim 61231, Germany
| | - Judith Magenheim
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - David M Barry
- Department of Molecular Biology and Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Simon Lee
- Department of Molecular Biology and Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Leilani Marty-Santos
- Program in Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Stephen Fu
- Department of Molecular Biology and Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Yuval Dor
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Ondine Cleaver
- Department of Molecular Biology and Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA.
| |
Collapse
|
29
|
Dong W, Xian Y, Yuan W, Huifeng Z, Tao W, Zhiqiang L, Shan F, Ya F, Hongli W, Jinghuan W, Lei Q, Li Z, Hongyi Q. Catalpol stimulates VEGF production via the JAK2/STAT3 pathway to improve angiogenesis in rats' stroke model. JOURNAL OF ETHNOPHARMACOLOGY 2016; 191:169-179. [PMID: 27301615 DOI: 10.1016/j.jep.2016.06.030] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 06/05/2016] [Accepted: 06/08/2016] [Indexed: 05/25/2023]
Abstract
ETHNOBOTANICAL RELEVANCE Catalpol is the main active component of the radix from Rehmannia glutinosa Libosch, which has pleiotropic protective effects in neurodegenerative diseases, ischemic stroke, metabolic disorders and others AIM Catalpol has been shown to have neuroprotective, neurorepair, and angiogenesis effects following ischemic brain injury. However, its molecular mechanisms are still poorly understood. In previous studies, the JAK2/STAT3 signaling pathway was found to play a role in neuroprotection and angiogenesis. This study investigated the role of catalpol in stimulating angiogenesis via the JAK2/STAT3 pathway after permanent focal cerebral ischemia (pMCAO). METHODS Rats were subjected to right middle cerebral artery occlusion through electrocoagulation and were treated with catalpol (5mg/kg), AG490 was also used to inhibit STAT3 phosphorylation (pSTAT3). RESULTS Following stroke, Catalpol improved the neuroethology deficit, increased the cerebral blood flow (CBF) of infarcted brain and upregulated EPO and EPOR. AG490 suppressed the phosphorylation of signal transducer and activator of transcription 3 (STAT3), ultimately inhibited VEGF mRNA expression, which reduced VEGF protein expression and inhibited stroke-induced angiogenesis. However, Catalpol enhanced stroke-induced STAT3 activation and subsequently restored STAT3 activity through the recovery of STAT3 binding to VEGF. Moreover, Catalpol reversed the effect of AG490 on STAT3 activation and nuclear translocation, restored the transcriptional activity of the VEGF promoter by recruiting STAT3 to the VEGF promoter, improved VEGF mRNA and protein expression, increased angiogenesis, reduced the difference in CBF between the infarcted and intact brain and ameliorated the neuroethology behaviors after stroke. CONCLUSION Catalpol affects neuroprotection and angiogenesis via the JAK2/STAT3 signaling pathway, which is mediated by STAT3 activation and VEGF expression. Catalpol may be used as a potential therapeutic drug for stroke.
Collapse
MESH Headings
- Angiogenesis Inducing Agents/pharmacology
- Animals
- Brain/drug effects
- Brain/enzymology
- Brain/pathology
- Brain/physiopathology
- Cerebral Arteries/drug effects
- Cerebral Arteries/enzymology
- Cerebral Arteries/pathology
- Cerebral Arteries/physiopathology
- Cerebrovascular Circulation/drug effects
- Disease Models, Animal
- Erythropoietin/metabolism
- Infarction, Middle Cerebral Artery/drug therapy
- Infarction, Middle Cerebral Artery/enzymology
- Infarction, Middle Cerebral Artery/pathology
- Infarction, Middle Cerebral Artery/physiopathology
- Iridoid Glucosides/pharmacology
- Janus Kinase 2/metabolism
- Male
- Neovascularization, Physiologic/drug effects
- Neuroprotective Agents/pharmacology
- Phosphorylation
- Promoter Regions, Genetic
- Protein Binding
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Sprague-Dawley
- Receptors, Erythropoietin/metabolism
- STAT3 Transcription Factor/metabolism
- Signal Transduction/drug effects
- Time Factors
- Transcriptional Activation
- Up-Regulation
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Wan Dong
- Department of Emergency, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yang Xian
- Department of Pharmacy, The Seventh People's Hospital of Chengdu, Chengdu 610041, China
| | - Wang Yuan
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China; Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Zhu Huifeng
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China; Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China.
| | - Wang Tao
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China; Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Liu Zhiqiang
- Department of Pharmacy, The First People's Hospital of Neijiang, Neijiang 641000, China
| | - Feng Shan
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China; Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Fu Ya
- College of Chemistry and Chemical Engineering, Chongqing University of Science & Technology, Chongqing 401331, China
| | - Wang Hongli
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China; Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Wang Jinghuan
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China; Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Qin Lei
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China; Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Zou Li
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China; Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Qi Hongyi
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China; Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| |
Collapse
|
30
|
Boulay AC, Cisternino S, Cohen-Salmon M. Immunoregulation at the gliovascular unit in the healthy brain: A focus on Connexin 43. Brain Behav Immun 2016; 56:1-9. [PMID: 26674996 DOI: 10.1016/j.bbi.2015.11.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 10/26/2015] [Accepted: 11/28/2015] [Indexed: 01/18/2023] Open
Abstract
In the brain, immune cell infiltration is normally kept at a very low level and a unique microenvironment strictly restricts immune reactions and inflammation. Even in such quiescent environment, a constant immune surveillance is at work allowing the brain to rapidly react to threats. To date, knowledge about the factors regulating the brain-immune system interrelationship in healthy conditions remains elusive. Interestingly, astrocytes, the most abundant glial cells in the brain, may participate in many aspects of this unique homeostasis, in particular due to their close interaction with the brain vascular system and expression of a specific molecular repertoire. Indeed, astrocytes maintain the blood-brain barrier (BBB) integrity, interact with immune cells, and participate in the regulation of intracerebral liquid movements. We recently showed that Connexin 43 (Cx43), a gap junction protein highly expressed by astrocytes at the BBB interface, is an immunoregulating factor. The absence of astroglial Cx43 leads to a transient endothelial activation, a continuous immune recruitment as well as the development of a specific humoral autoimmune response against the von Willebrand factor A domain-containing protein 5a, an extracellular matrix protein expressed by astrocytes. In this review, we propose to gather current knowledge on how astrocytes may influence the immune system in the healthy brain, focusing on their roles at the gliovascular interface. We will also consider pathological situations involving astrocyte-specific autoimmunities. Finally, we will discuss the specific role of astroglial Cx43 and the physiological consequences of immune regulations taking place on inflammation, cognition and behavior in the absence of Cx43.
Collapse
Affiliation(s)
- Anne-Cécile Boulay
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Centre National de la Recherche Scientifique CNRS, Unité Mixte de Recherche 7241, Institut National de la Santé et de la Recherche Médicale INSERM, U1050, Neuroglial Interactions in Cerebral Physiopathology, 75231 Paris Cedex 05, France; University Pierre et Marie Curie, ED, N°158, 75005 Paris, France; MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, 75005 Paris, France
| | - Salvatore Cisternino
- Variabilité de réponse aux psychotropes, INSERM, U1144, Paris F-75006, France; Université Paris Descartes, Faculté de Pharmacie, UMR-S 1144, 75006 Paris, France; Université Paris Diderot, UMR-S 1144, 75013 Paris, France
| | - Martine Cohen-Salmon
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Centre National de la Recherche Scientifique CNRS, Unité Mixte de Recherche 7241, Institut National de la Santé et de la Recherche Médicale INSERM, U1050, Neuroglial Interactions in Cerebral Physiopathology, 75231 Paris Cedex 05, France; University Pierre et Marie Curie, ED, N°158, 75005 Paris, France; MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, 75005 Paris, France.
| |
Collapse
|
31
|
Hao W, Friedman A. Serum uPAR as Biomarker in Breast Cancer Recurrence: A Mathematical Model. PLoS One 2016; 11:e0153508. [PMID: 27078836 PMCID: PMC4831695 DOI: 10.1371/journal.pone.0153508] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/30/2016] [Indexed: 12/22/2022] Open
Abstract
There are currently over 2.5 million breast cancer survivors in the United States and, according to the American Cancer Society, 10 to 20 percent of these women will develop recurrent breast cancer. Early detection of recurrence can avoid unnecessary radical treatment. However, self-examination or mammography screening may not discover a recurring cancer if the number of surviving cancer cells is small, while biopsy is too invasive and cannot be frequently repeated. It is therefore important to identify non-invasive biomarkers that can detect early recurrence. The present paper develops a mathematical model of cancer recurrence. The model, based on a system of partial differential equations, focuses on tissue biomarkers that include the plasminogen system. Among them, only uPAR is known to have significant correlation to its concentration in serum and could therefore be a good candidate for serum biomarker. The model includes uPAR and other associated cytokines and cells. It is assumed that the residual cancer cells that survived primary cancer therapy are concentrated in the same location within a region with a very small diameter. Model simulations establish a quantitative relation between the diameter of the growing cancer and the total uPAR mass in the cancer. This relation is used to identify uPAR as a potential serum biomarker for breast cancer recurrence.
Collapse
Affiliation(s)
- Wenrui Hao
- Mathematical Biosciences Institute, The Ohio State University, Columbus, OH, United States of America
| | - Avner Friedman
- Mathematical Biosciences Institute, The Ohio State University, Columbus, OH, United States of America
- Department of Mathematics, The Ohio State University, Columbus, OH, United States of America
| |
Collapse
|
32
|
Batarseh YS, Duong QV, Mousa YM, Al Rihani SB, Elfakhri K, Kaddoumi A. Amyloid-β and Astrocytes Interplay in Amyloid-β Related Disorders. Int J Mol Sci 2016; 17:338. [PMID: 26959008 PMCID: PMC4813200 DOI: 10.3390/ijms17030338] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 02/14/2016] [Accepted: 02/24/2016] [Indexed: 12/17/2022] Open
Abstract
Amyloid-β (Aβ) pathology is known to promote chronic inflammatory responses in the brain. It was thought previously that Aβ is only associated with Alzheimer's disease and Down syndrome. However, studies have shown its involvement in many other neurological disorders. The role of astrocytes in handling the excess levels of Aβ has been highlighted in the literature. Astrocytes have a distinctive function in both neuronal support and protection, thus its involvement in Aβ pathological process may tip the balance toward chronic inflammation and neuronal death. In this review we describe the involvement of astrocytes in Aβ related disorders including Alzheimer's disease, Down syndrome, cerebral amyloid angiopathy, and frontotemporal dementia.
Collapse
Affiliation(s)
- Yazan S Batarseh
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 70504, USA.
| | - Quoc-Viet Duong
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 70504, USA.
| | - Youssef M Mousa
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 70504, USA.
| | - Sweilem B Al Rihani
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 70504, USA.
| | - Khaled Elfakhri
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 70504, USA.
| | - Amal Kaddoumi
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 70504, USA.
| |
Collapse
|
33
|
Abeysinghe HCS, Phillips EL, Chin-Cheng H, Beart PM, Roulston CL. Modulating Astrocyte Transition after Stroke to Promote Brain Rescue and Functional Recovery: Emerging Targets Include Rho Kinase. Int J Mol Sci 2016; 17:288. [PMID: 26927079 PMCID: PMC4813152 DOI: 10.3390/ijms17030288] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/26/2016] [Accepted: 02/05/2016] [Indexed: 01/13/2023] Open
Abstract
Stroke is a common and serious condition, with few therapies. Whilst previous focus has been directed towards biochemical events within neurons, none have successfully prevented the progression of injury that occurs in the acute phase. New targeted treatments that promote recovery after stroke might be a better strategy and are desperately needed for the majority of stroke survivors. Cells comprising the neurovascular unit, including blood vessels and astrocytes, present an alternative target for supporting brain rescue and recovery in the late phase of stroke, since alteration in the unit also occurs in regions outside of the lesion. One of the major changes in the unit involves extensive morphological transition of astrocytes resulting in altered energy metabolism, decreased glutamate reuptake and recycling, and retraction of astrocyte end feed from both blood vessels and neurons. Whilst globally inhibiting transitional change in astrocytes after stroke is reported to result in further damage and functional loss, we discuss the available evidence to suggest that the transitional activation of astrocytes after stroke can be modulated for improved outcomes. In particular, we review the role of Rho-kinase (ROCK) in reactive gliosis and show that inhibiting ROCK after stroke results in reduced scar formation and improved functional recovery.
Collapse
Affiliation(s)
- Hima Charika S Abeysinghe
- Neurotrauma Research, Department of Medicine, St Vincent's Campus, University of Melbourne, Parkville, VIC 3065, Australia.
- Department of Surgery, St Vincent's Campus, University of Melbourne, Parkville, VIC 3065, Australia.
| | - Ellie L Phillips
- Department of Biochemistry and Molecular Biology, Bio21 Insitute, University of Melbourne, Parkville, VIC 3010, Australia.
| | - Heung Chin-Cheng
- Department of Biochemistry and Molecular Biology, Bio21 Insitute, University of Melbourne, Parkville, VIC 3010, Australia.
| | - Philip M Beart
- The Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, Parkville, VIC 3010, Australia.
| | - Carli L Roulston
- Neurotrauma Research, Department of Medicine, St Vincent's Campus, University of Melbourne, Parkville, VIC 3065, Australia.
| |
Collapse
|
34
|
Bag AK, Kim H, Gao Y, Bolding M, Warren PP, Fathallah-Shaykh HM, Gurler D, Markert JM, Fiveash J, Beasley TM, Khawaja A, Friedman GK, Chapman PR, Nabors LB, Han X. Prolonged treatment with bevacizumab is associated with brain atrophy: a pilot study in patients with high-grade gliomas. J Neurooncol 2015; 122:585-93. [PMID: 25711673 DOI: 10.1007/s11060-015-1751-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 02/17/2015] [Indexed: 12/17/2022]
Abstract
Bevacizumab is widely used for treatment of high-grade gliomas and other malignancies. Because bevacizumab has been shown to be associated with neurocognitive decline, this study is designed to investigate whether prolonged treatment with bevacizumab is also associated with brain atrophy. We identified 12 high-grade glioma patients who received bevacizumab for 12 months at the first recurrence and 13 matched controls and blindly compared the volumes of the contralateral hemispheres and contralateral ventricle in these two groups at baseline and after 12 ± 2 months of the baseline scan by two independent analyses. The volumes of the contralateral hemispheres and ventricles did not differ significantly between the two groups at baseline. Whereas, in the control group the volumes of the contralateral hemisphere changed subtly from baseline to follow-up (p = 0.23), in the bevacizumab-treated group the volumes significantly decreased from baseline to follow-up (p = 0.03). There was significant increase in the contralateral ventricle volume from base line to follow-up scans in both the control group (p = 0.01) and in the bevacizumab group (p = 0.005). Both the absolute and the percentage changes of contralateral hemisphere volumes and contralateral ventricular volumes between the two patient groups were statistically significant (p < 0.05). Results of this study demonstrate prolonged treatment with bevacizumab is associated with atrophy of the contralateral brain hemisphere.
Collapse
Affiliation(s)
- Asim K Bag
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Nielsen CM, Cuervo H, Ding VW, Kong Y, Huang EJ, Wang RA. Deletion of Rbpj from postnatal endothelium leads to abnormal arteriovenous shunting in mice. Development 2014; 141:3782-92. [PMID: 25209249 DOI: 10.1242/dev.108951] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Arteriovenous malformations (AVMs) are tortuous vessels characterized by arteriovenous (AV) shunts, which displace capillaries and shunt blood directly from artery to vein. Notch signaling regulates embryonic AV specification by promoting arterial, as opposed to venous, endothelial cell (EC) fate. To understand the essential role of endothelial Notch signaling in postnatal AV organization, we used inducible Cre-loxP recombination to delete Rbpj, a mediator of canonical Notch signaling, from postnatal ECs in mice. Deletion of endothelial Rbpj from birth resulted in features of AVMs by P14, including abnormal AV shunting and tortuous vessels in the brain, intestine and heart. We further analyzed brain AVMs, as they pose particular health risks. Consistent with AVM pathology, we found cerebral hemorrhage, hypoxia and necrosis, and neurological deficits. AV shunts originated from capillaries (and possibly venules), with the earliest detectable morphological abnormalities in AV connections by P8. Prior to AV shunt formation, alterations in EC gene expression were detected, including decreased Efnb2 and increased Pai1, which encodes a downstream effector of TGFβ signaling. After AV shunts had formed, whole-mount immunostaining showed decreased Efnb2 and increased Ephb4 expression within AV shunts, suggesting that ECs were reprogrammed from arterial to venous identity. Deletion of Rbpj from adult ECs led to tortuosities in gastrointestinal, uterine and skin vascular beds, but had mild effects in the brain. Our results demonstrate a temporal requirement for Rbpj in postnatal ECs to maintain proper artery, capillary and vein organization and to prevent abnormal AV shunting and AVM pathogenesis.
Collapse
Affiliation(s)
- Corinne M Nielsen
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Henar Cuervo
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Vivianne W Ding
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Yupeng Kong
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Eric J Huang
- Department of Pathology, University of California, San Francisco, CA 94143, USA
| | - Rong A Wang
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
36
|
Vascular endothelial growth factor increases during blood-brain barrier-enhanced permeability caused by Phoneutria nigriventer spider venom. BIOMED RESEARCH INTERNATIONAL 2014; 2014:721968. [PMID: 25247186 PMCID: PMC4163422 DOI: 10.1155/2014/721968] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Revised: 07/26/2014] [Accepted: 08/05/2014] [Indexed: 11/17/2022]
Abstract
Phoneutria nigriventer spider accidental envenomation provokes neurotoxic manifestations, which when critical, results in epileptic-like episodes. In rats, P. nigriventer venom (PNV) causes blood-brain barrier breakdown (BBBb). The PNV-induced excitotoxicity results from disturbances on Na+, K+ and Ca2+ channels and glutamate handling. The vascular endothelial growth factor (VEGF), beyond its angiogenic effect, also, interferes on synaptic physiology by affecting the same ion channels and protects neurons from excitotoxicity. However, it is unknown whether VEGF expression is altered following PNV envenomation. We found that adult and neonates rats injected with PNV showed immediate neurotoxic manifestations which paralleled with endothelial occludin, β-catenin, and laminin downregulation indicative of BBBb. In neonate rats, VEGF, VEGF mRNA, and Flt-1 receptors, glutamate decarboxylase, and calbindin-D28k increased in Purkinje neurons, while, in adult rats, the BBBb paralleled with VEGF mRNA, Flk-1, and calbindin-D28k increases and Flt-1 decreases. Statistically, the variable age had a role in such differences, which might be due to age-related unequal maturation of blood-brain barrier (BBB) and thus differential cross-signaling among components of the glial neurovascular unit. The concurrent increases in the VEGF/Flt-1/Flk-1 system in the cerebellar neuron cells and the BBBb following PNV exposure might imply a cytokine modulation of neuronal excitability consequent to homeostatic perturbations induced by ion channels-acting PNV neuropeptides. Whether such modulation represents neuroprotection needs further investigation.
Collapse
|
37
|
Delayed VEGF treatment enhances angiogenesis and recovery after neonatal focal rodent stroke. Transl Stroke Res 2014; 4:189-200. [PMID: 23926451 DOI: 10.1007/s12975-012-0221-6] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Neonatal stroke occurs in one in 4,000 live births and leads to significant morbidity and mortality. Approximately two thirds of the survivors have long-term sequelae including seizures and neurological deficits. However, the pathophysiological mechanisms of recovery after neonatal stroke are not clearly understood, and preventive measures and treatments are nonexistent in the clinical setting. In this study, we investigated the effect of vascular endothelial growth factor (VEGF) treatment on histological recovery and angiogenic response to the developing brain after an ischemic insult. Ten-day-old Sprague-Dawley rats underwent right middle cerebral arterial occlusion (MCAO) for 1.5 h. Diffusion-weighted MRI during occlusion confirmed focal ischemia that was then followed by reperfusion. On group of animals received 5-bromo-2-deoxyuridine and sacrificed at postnatal day (P)18 or P25. A second group of animals was treated with VEGF (1.5 µg/kg, icv) or phosphate-buffered saline (PBS) at P18 and perfusion fixed at P25. Based on Nissl and iron staining, a single VEGF injection reduced the injury score, compared to the animals that underwent MCAO and PBS injection. Furthermore, neurodegeneration represented by neuronal nuclei staining was markedly diminished. In addition, animals treated with VEGF revealed a positive trend in endothelial proliferation and a significant increase in total vessel volume in the peri-infarct region of the caudate. The number of Iba1-positive microglial cells was significantly reduced after a single VEGF injection, and myelin basic protein expression was enhanced in the caudate after ischemia without an effect of VEGF treatment. In conclusion, delayed treatment with VEGF ameliorates injury, promotes endothelial cell proliferation, and increases total vascular volume following neonatal stroke. These results suggest that VEGF has a neuroprotective effect, in part by enhancing endogenous angiogenesis. These data contribute to a better understanding of neonatal stroke.
Collapse
|
38
|
No changes in cerebellar microvessel length density in sudden infant death syndrome: implications for pathogenetic mechanisms. J Neuropathol Exp Neurol 2014; 73:312-23. [PMID: 24607967 DOI: 10.1097/nen.0000000000000055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Sudden infant death syndrome (SIDS) is the leading cause of mortality in infants younger than 1 year in developed countries, but its primary cause remains unknown. Some studies suggest that there may be hypoxia in the cerebellum in SIDS subjects, but mean total Purkinje cell numbers in SIDS versus controls was recently found not to be different. Probably the best marker for chronic hypoxia in a brain region is the microvessel length per unit volume of tissue, that is, the microvessel length density (MLD). Here, we investigated MLDs using a rigorous design-based stereologic approach in all cell layers and white matter in postmortem cerebella from 9 SIDS cases who died between ages 2 and 10 months and from 14 control children, 9 of which were age- and sex- matched to the SIDS cases. We found no differences either in mean MLDs in the cerebellar layers between the SIDS cases and the controls or between controls with a low likelihood of hypoxia and those with a higher likelihood of hypoxia. Immunohistochemical detection of the astrocytosis marker glial fibrillary acidic protein showed no differences between the SIDS and the matched control cases. These data indicate that there is no association of chronic hypoxia in the cerebellum with SIDS.
Collapse
|
39
|
Ward NL, Lamanna JC. The neurovascular unit and its growth factors: coordinated response in the vascular and nervous systems. Neurol Res 2013; 26:870-83. [PMID: 15727271 DOI: 10.1179/016164104x3798] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The nervous and vascular systems contain many common organizational features and develop similarly in terms of anatomical patterning. During embryogenesis and in regions of the brain undergoing postnatal neurogenesis, neural stem cells and endothelial cells are found in close proximity, or within a so-called vascular niche. The similarities in patterning and proximity may reflect coordinated development based on responsiveness to similar growth factors such as vascular endothelial growth factor, semaphorin, and ephrins/Ephs: molecules involved in the development and maintenance of both the nervous and vascular systems. Despite the blatant similarities between the vascular and nervous systems, little is still known about the co-dependence and/or interactions between the two systems during development and following alterations in metabolic demand as seen during aging, exercise, and disease processes. The interactions between the two systems involving common growth factors suggest these two systems have evolved in an interconnected way.
Collapse
Affiliation(s)
- Nicole L Ward
- Department of Anatomy, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
| | | |
Collapse
|
40
|
Licht T, Keshet E. Delineating multiple functions of VEGF-A in the adult brain. Cell Mol Life Sci 2013; 70:1727-37. [PMID: 23475068 PMCID: PMC11113886 DOI: 10.1007/s00018-013-1280-x] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 01/28/2013] [Accepted: 01/28/2013] [Indexed: 02/07/2023]
Abstract
Vascular endothelial growth factor-A (abbreviated throughout this review as VEGF) is mostly known for its angiogenic activity, for its activity as a vascular permeability factor, and for its vascular survival activity [1]. There is a growing body of evidence, however, that VEGF fulfills additional less 'traditional' functions in multiple organs, both during development, as well as homeostatic functions in fully developed organs. This review focuses on the multiple roles of VEGF in the adult brain and is less concerned with the roles played by VEGF during brain development, functions described elsewhere in this review series. Most functions of VEGF that are essential for proper brain development are, in fact, dispensable in the adult brain as was clearly demonstrated using a conditional brain-specific VEGF loss-of-function (LOF) approach. Thus, in contrast to VEGF LOF in the developing brain, a process which is detrimental for the growth and survival of blood vessels and leads to massive neuronal apoptosis [2-4], continued signaling by VEGF in the mature brain is no longer required for maintaining already established cerebral vasculature and its inhibition does not cause appreciable vessel regression, hypoxia or apoptosis [4-7]. Yet, VEGF continues to be expressed in the adult brain in a constitutive manner. Moreover, VEGF is expressed in the adult brain in a region-specific manner and in distinctive spatial patterns incompatible with an angiogenic role (see below), strongly suggesting angiogenesis-independent and possibly also perfusion-independent functions. Here we review current knowledge on some of these 'non-traditional', often unexpected homeostatic VEGF functions, including those unrelated to its effects on the brain vasculature. These effects could be mediated directly (on non-vascular cells expressing cognate VEGF receptors) or indirectly (via the endothelium). Experimental approaches aimed at distinguishing between these possibilities for each particular VEGF function will be described. This review is only concerned with homeostatic functions of VEGF in the normal, non-injured brain. The reader is referred elsewhere in this series for a review on VEGF actions in response to various forms of brain injury and/or brain pathology.
Collapse
Affiliation(s)
- Tamar Licht
- Department of Developmental Biology and Cancer Research, Hadassah Medical School, The Hebrew University, Jerusalem, Israel
| | - Eli Keshet
- Department of Developmental Biology and Cancer Research, Hadassah Medical School, The Hebrew University, Jerusalem, Israel
| |
Collapse
|
41
|
Kundi S, Bicknell R, Ahmed Z. The role of angiogenic and wound-healing factors after spinal cord injury in mammals. Neurosci Res 2013; 76:1-9. [PMID: 23562792 DOI: 10.1016/j.neures.2013.03.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 03/18/2013] [Accepted: 03/20/2013] [Indexed: 12/17/2022]
Abstract
Patients with spinal cord injury (SCI) are permanently paralysed and anaesthetic below the lesion. This morbidity is attributed to the deposition of a dense scar at the injury site, the cellular components of which secrete axon growth inhibitory ligands that prevent severed axons reconnecting with denervated targets. Another complication of SCI is wound cavitation where a fluid filled cyst forms in the peri-lesion neuropil, enlarging over the first few months after injury and causes secondary axonal damage. Wound healing after SCI is accompanied by angiogenesis, which is regulated by angiogenic proteins, produced in response to oxygen deprivation. Necrosis in and about the SCI lesion sites may be suppressed by promoting angiogenesis and the resulting neuropil protection will enhance recovery after SCI. This review addresses the use of angiogenic/wound-healing related proteins including vascular endothelial growth factor, fibroblast growth factor, angiopoietin-1, angiopoietin-2 and transforming growth factor-β to moderate necrosis and axon sparing after SCI, providing a conducive environment for growth essential to functional recovery.
Collapse
Affiliation(s)
- Sarina Kundi
- Neurotrauma and Neurodegeneration, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, UK
| | | | | |
Collapse
|
42
|
Wittko-Schneider IM, Schneider FT, Plate KH. Brain homeostasis: VEGF receptor 1 and 2-two unequal brothers in mind. Cell Mol Life Sci 2013; 70:1705-25. [PMID: 23475067 PMCID: PMC3632714 DOI: 10.1007/s00018-013-1279-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 01/28/2013] [Accepted: 01/28/2013] [Indexed: 12/15/2022]
Abstract
Vascular endothelial growth factors (VEGFs), initially thought to act specifically on the vascular system, exert trophic effects on neural cells during development and adulthood. Therefore, the VEGF system serves as a promising therapeutic target for brain pathologies, but its simultaneous action on vascular cells paves the way for harmful side effects. To circumvent these deleterious effects, many studies have aimed to clarify whether VEGFs directly affect neural cells or if the effects are mediated secondarily via other cell types, like vascular cells. A great number of reports have shown the expression and function of VEGF receptors (VEGFRs), mainly VEGFR-1 and -2, in neural cells, where VEGFR-2 has been described as the major mediator of VEGF-A signals. This review aims to summarize and compare the divergent roles of VEGFR-1 and -2 during CNS development and homeostasis.
Collapse
Affiliation(s)
- Ina M Wittko-Schneider
- Neuroscience Center, Institute of Neurology (Edinger Institute), Goethe University Medical School, Heinrich-Hoffmann Strasse 7, 60528, Frankfurt, Germany.
| | | | | |
Collapse
|
43
|
Udan RS, Culver JC, Dickinson ME. Understanding vascular development. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2012; 2:327-46. [PMID: 23799579 DOI: 10.1002/wdev.91] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The vasculature of an organism has the daunting task of connecting all the organ systems to nourish tissue and sustain life. This complex network of vessels and associated cells must maintain blood flow, but constantly adapt to acute and chronic changes within tissues. While the vasculature has been studied for over a century, we are just beginning to understand the processes that regulate its formation and how genetic hierarchies are influenced by mechanical and metabolic cues to refine vessel structure and optimize efficiency. As we gain insights into the developmental mechanisms, it is clear that the processes that regulate blood vessel development can also enable the adult to adapt to changes in tissues that can be elicited by exercise, aging, injury, or pathology. Thus, research in vessel development has provided tremendous insights into therapies for vascular diseases and disorders, cancer interventions, wound repair and tissue engineering, and in turn, these models have clearly impacted our understanding of development. Here we provide an overview of the development of the vascular system, highlighting several areas of active investigation and key questions that remain to be answered.
Collapse
Affiliation(s)
- Ryan S Udan
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | | | | |
Collapse
|
44
|
Szomolay B, Eubank TD, Roberts RD, Marsh CB, Friedman A. Modeling the inhibition of breast cancer growth by GM-CSF. J Theor Biol 2012; 303:141-51. [PMID: 22763136 DOI: 10.1016/j.jtbi.2012.03.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Revised: 12/30/2011] [Accepted: 03/18/2012] [Indexed: 12/23/2022]
Abstract
M-CSF is overexpressed in breast cancer and is known to stimulate macrophages to produce VEGF resulting in angiogenesis. It has recently been shown that the growth factor GM-CSF injected into murine breast tumors slowed tumor growth by secreting soluble VEGF receptor-1 (sVEGFR-1) that binds and inactivates VEGF. This study presents a mathematical model that includes all the components above, as well as MCP-1, tumor cells, and oxygen. The model simulations are representative of the in vivo data through predictions of tumor growth using different protocol strategies for GM-CSF for the purpose of predicting higher degrees of treatment success. For example, our model predicts that once a week dosing of GM-CSF would be less effective than daily, twice a week, or three times a week treatment because of the presence of essential factors required for the anti-tumor effect of GM-CSF.
Collapse
Affiliation(s)
- Barbara Szomolay
- Mathematical Biosciences Institute, The Ohio State University, USA.
| | | | | | | | | |
Collapse
|
45
|
Cvetanovic M, Patel JM, Marti HH, Kini AR, Opal P. Vascular endothelial growth factor ameliorates the ataxic phenotype in a mouse model of spinocerebellar ataxia type 1. Nat Med 2011; 17:1445-7. [PMID: 22001907 PMCID: PMC3287040 DOI: 10.1038/nm.2494] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Accepted: 08/24/2011] [Indexed: 11/15/2022]
Abstract
SCA1 is an adult-onset, dominantly inherited neurodegenerative disease caused by expansion of a glutamine repeat tract in ATXN1. Although the precise function of ATXN1 remains elusive, it appears to play a role in transcriptional repression. We find that mutant ATXN1 suppresses transcription of the neurotrophic and angiogenic factor VEGF. We also show that genetic or pharmacologic replenishment of VEGF mitigates SCA1 pathogenesis, suggesting a novel therapeutic strategy for this incurable disease.
Collapse
Affiliation(s)
- Marija Cvetanovic
- Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | | | | | | | | |
Collapse
|
46
|
Durham-Lee JC, Wu Y, Mokkapati VUL, Paulucci-Holthauzen AA, Nesic O. Induction of angiopoietin-2 after spinal cord injury. Neuroscience 2011; 202:454-64. [PMID: 22020092 DOI: 10.1016/j.neuroscience.2011.09.058] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 09/22/2011] [Accepted: 09/22/2011] [Indexed: 01/27/2023]
Abstract
Angiopoietin-1 (Ang-1) and angiopoietin-2 (Ang-2) have opposing effects on blood vessels, with Ang-2 being mainly induced during the endothelial barrier breakdown. It is known that spinal cord injury (SCI) induces lasting decreases in Ang-1 levels, underlying endothelial barrier disruption, but the expression of Ang-2 in spinal cord injury has not been studied. We characterized Ang-2 after SCI using a clinically relevant rat model of contusion SCI. We found that SCI induces marked and persistent upregulation of Ang-2 (up to 10 weeks after SCI), which does not reflect well-characterized temporal profile of the blood-spinal cord barrier (BSCB) breakdown after SCI, and thus suggests other role(s) for Ang-2 in injured spinal cords. Furthermore, we also found that higher Ang-2 levels were associated with more successful locomotor recovery after SCI, both in SCI rats with markedly better spontaneous motor recovery and in SCI rats receiving a neuroprotective pharmacological intervention (amiloride), suggesting a beneficial role for Ang-2 in injured spinal cords. Immunocytochemical analyses revealed that Ang-2 was not induced in endothelial cells, but in perivascular and non-vascular cells labeled with glial fibrillary acidic protein (GFAP) or with chondroitin sulfate proteoglycan (NG2). Therefore, it is unlikely that induction of Ang-2 contributes to vascular dysfunction underlying functional impairment after SCI, but rather that it contributes to the beneficial pro-angiogenic and/or gliogenic processes underlying recovery processes after SCI.
Collapse
Affiliation(s)
- J C Durham-Lee
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555-1072, USA
| | | | | | | | | |
Collapse
|
47
|
Boulday G, Rudini N, Maddaluno L, Blécon A, Arnould M, Gaudric A, Chapon F, Adams RH, Dejana E, Tournier-Lasserve E. Developmental timing of CCM2 loss influences cerebral cavernous malformations in mice. ACTA ACUST UNITED AC 2011; 208:1835-47. [PMID: 21859843 PMCID: PMC3171098 DOI: 10.1084/jem.20110571] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
As revealed in a new model of cerebral cavernous malformations (CCM), the timing of ablation of Ccm genes determines whether or not CCM lesions arise in brain and retina venous beds. Cerebral cavernous malformations (CCM) are vascular malformations of the central nervous system (CNS) that lead to cerebral hemorrhages. Familial CCM occurs as an autosomal dominant condition caused by loss-of-function mutations in one of the three CCM genes. Constitutive or tissue-specific ablation of any of the Ccm genes in mice previously established the crucial role of Ccm gene expression in endothelial cells for proper angiogenesis. However, embryonic lethality precluded the development of relevant CCM mouse models. Here, we show that endothelial-specific Ccm2 deletion at postnatal day 1 (P1) in mice results in vascular lesions mimicking human CCM lesions. Consistent with CCM1/3 involvement in the same human disease, deletion of Ccm1/3 at P1 in mice results in similar CCM lesions. The lesions are located in the cerebellum and the retina, two organs undergoing intense postnatal angiogenesis. Despite a pan-endothelial Ccm2 deletion, CCM lesions are restricted to the venous bed. Notably, the consequences of Ccm2 loss depend on the developmental timing of Ccm2 ablation. This work provides a highly penetrant and relevant CCM mouse model.
Collapse
Affiliation(s)
- Gwénola Boulday
- Institut National de la Santé et de la Recherche Médicale, UMR-S 740, 75010 Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Dale-Nagle EA, Satriotomo I, Mitchell GS. Spinal vascular endothelial growth factor induces phrenic motor facilitation via extracellular signal-regulated kinase and Akt signaling. J Neurosci 2011; 31:7682-90. [PMID: 21613481 PMCID: PMC3172810 DOI: 10.1523/jneurosci.0239-11.2011] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 03/26/2011] [Accepted: 03/31/2011] [Indexed: 12/21/2022] Open
Abstract
Although vascular endothelial growth factor (VEGFA-165) is primarily known for its role in angiogenesis, it also plays important neurotrophic and neuroprotective roles for spinal motor neurons. VEGFA-165 signals by activating its receptor tyrosine kinase VEGF receptor-2 (VEGFR-2). Because another growth/trophic factor that signals via a receptor tyrosine kinase (brain derived neurotrophic factor) elicits a long-lasting facilitation of respiratory motor activity in the phrenic nerve, we tested the hypothesis that VEGFA-165 elicits similar phrenic motor facilitation (pMF). Using immunohistochemistry and retrograde labeling techniques, we demonstrate that VEGFA-165 and VEGFR-2 are expressed in identified phrenic motor neurons. Furthermore, intrathecal VEGFA-165 administration at C4 elicits long-lasting pMF; intraspinal VEGFA-165 increased integrated phrenic nerve burst amplitude for at least 90 min after injection (53.1 ± 5.0% at 90 min; p < 0.001). Intrathecal VEGFA-165 increased phosphorylation (and presumed activation) of signaling molecules downstream from VEGFR-2 within the phrenic motor nucleus, including ERK (1.53 ± 0.13 vs 1.0 ± 0.05 arbitrary units in control rats; p < 0.05) and Akt (2.16 ± 0.41 vs 1.0 ± 0.41 arbitrary units in control rats; p < 0.05). VEGF-induced pMF was attenuated by the MEK/ERK inhibitor U0126 [1,4-diamino-2,3-dicyano-1,4-bis(o-aminophenylmercapto)butadiene] and was abolished by the phosphotidinositol 3 kinase/Akt inhibitor LY294002 [2-(4-morpholinyl)-8-phenyl-1(4H)-benzopyran-4-one hydrochloride], demonstrating that ERK mitogen-activated protein kinases and Akt are both required for full expression of VEGF-induced pMF. This is the first report that VEGFA-165 elicits plasticity in any motor system. Furthermore, because VEGFA-165 expression is hypoxia sensitive, it may play a role in respiratory plasticity after prolonged exposures to low oxygen.
Collapse
Affiliation(s)
- Erica A. Dale-Nagle
- Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin 53706
| | - Irawan Satriotomo
- Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin 53706
| | - Gordon S. Mitchell
- Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin 53706
| |
Collapse
|
49
|
Calvo CF, Fontaine RH, Soueid J, Tammela T, Makinen T, Alfaro-Cervello C, Bonnaud F, Miguez A, Benhaim L, Xu Y, Barallobre MJ, Moutkine I, Lyytikkä J, Tatlisumak T, Pytowski B, Zalc B, Richardson W, Kessaris N, Garcia-Verdugo JM, Alitalo K, Eichmann A, Thomas JL. Vascular endothelial growth factor receptor 3 directly regulates murine neurogenesis. Genes Dev 2011; 25:831-44. [PMID: 21498572 PMCID: PMC3078708 DOI: 10.1101/gad.615311] [Citation(s) in RCA: 130] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Accepted: 02/24/2011] [Indexed: 12/25/2022]
Abstract
Neural stem cells (NSCs) are slowly dividing astrocytes that are intimately associated with capillary endothelial cells in the subventricular zone (SVZ) of the brain. Functionally, members of the vascular endothelial growth factor (VEGF) family can stimulate neurogenesis as well as angiogenesis, but it has been unclear whether they act directly via VEGF receptors (VEGFRs) expressed by neural cells, or indirectly via the release of growth factors from angiogenic capillaries. Here, we show that VEGFR-3, a receptor required for lymphangiogenesis, is expressed by NSCs and is directly required for neurogenesis. Vegfr3:YFP reporter mice show VEGFR-3 expression in multipotent NSCs, which are capable of self-renewal and are activated by the VEGFR-3 ligand VEGF-C in vitro. Overexpression of VEGF-C stimulates VEGFR-3-expressing NSCs and neurogenesis in the SVZ without affecting angiogenesis. Conversely, conditional deletion of Vegfr3 in neural cells, inducible deletion in subventricular astrocytes, and blocking of VEGFR-3 signaling with antibodies reduce SVZ neurogenesis. Therefore, VEGF-C/VEGFR-3 signaling acts directly on NSCs and regulates adult neurogenesis, opening potential approaches for treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Charles-Félix Calvo
- University Pierre et Marie Curie-Paris 6, Centre de Recherche de l'Institut du Cerveau et de la Moelle Epiniere, UMR S975, Paris 75651, France
- Inserm, U975, Paris 75651, France
- CNRS, UMR 7225, Paris 75651, France
| | - Romain H. Fontaine
- University Pierre et Marie Curie-Paris 6, Centre de Recherche de l'Institut du Cerveau et de la Moelle Epiniere, UMR S975, Paris 75651, France
- Inserm, U975, Paris 75651, France
- CNRS, UMR 7225, Paris 75651, France
| | - Jihane Soueid
- University Pierre et Marie Curie-Paris 6, Centre de Recherche de l'Institut du Cerveau et de la Moelle Epiniere, UMR S975, Paris 75651, France
- Inserm, U975, Paris 75651, France
- CNRS, UMR 7225, Paris 75651, France
| | - Tuomas Tammela
- Molecular/Cancer Biology Laboratory, Biomedicum Helsinki, University of Helsinki, Helsinki FI-00290, Finland
- Department of Pathology, Haartman Institute, University of Helsinki, Helsinki FI-00290, Finland
| | - Taija Makinen
- Molecular/Cancer Biology Laboratory, Biomedicum Helsinki, University of Helsinki, Helsinki FI-00290, Finland
- Department of Pathology, Haartman Institute, University of Helsinki, Helsinki FI-00290, Finland
| | - Clara Alfaro-Cervello
- Laboratorio de Morfología Celular, Centro de Investigación Príncipe Felipe, CIBERNED, Valencia 46013, Spain
| | - Fabien Bonnaud
- Cellular Imaging and Analysis, PerkinElmer, Coventry HP9 2FX, United Kingdom
| | - Andres Miguez
- University Pierre et Marie Curie-Paris 6, Centre de Recherche de l'Institut du Cerveau et de la Moelle Epiniere, UMR S975, Paris 75651, France
- Inserm, U975, Paris 75651, France
- CNRS, UMR 7225, Paris 75651, France
| | - Lucile Benhaim
- University Pierre et Marie Curie-Paris 6, Centre de Recherche de l'Institut du Cerveau et de la Moelle Epiniere, UMR S975, Paris 75651, France
- Inserm, U975, Paris 75651, France
- CNRS, UMR 7225, Paris 75651, France
| | - Yunling Xu
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, Paris 75005, France
| | - Maria-José Barallobre
- Center for Genomic Regulation, UPF and CIBER de Enfermedades Raras, Barcelona 08003, Spain
| | - Imane Moutkine
- University Pierre et Marie Curie-Paris 6, Centre de Recherche de l'Institut du Cerveau et de la Moelle Epiniere, UMR S975, Paris 75651, France
- Inserm, U975, Paris 75651, France
- CNRS, UMR 7225, Paris 75651, France
| | - Johannes Lyytikkä
- Experimental MRI Laboratory, Department of Neurology, Helsinki University Central Hospital, Helsinki FI-00290, Finland
| | - Turgut Tatlisumak
- Experimental MRI Laboratory, Department of Neurology, Helsinki University Central Hospital, Helsinki FI-00290, Finland
| | | | - Bernard Zalc
- University Pierre et Marie Curie-Paris 6, Centre de Recherche de l'Institut du Cerveau et de la Moelle Epiniere, UMR S975, Paris 75651, France
- Inserm, U975, Paris 75651, France
- CNRS, UMR 7225, Paris 75651, France
- Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris 75651, France
| | - William Richardson
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, United Kingdom
- Research Department of Cell and Developmental Biology, University College London, London WC1E 6BT, United Kingdom
| | - Nicoletta Kessaris
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, United Kingdom
- Research Department of Cell and Developmental Biology, University College London, London WC1E 6BT, United Kingdom
| | - Jose Manuel Garcia-Verdugo
- Laboratorio de Morfología Celular, Centro de Investigación Príncipe Felipe, CIBERNED, Valencia 46013, Spain
- Laboratorio de Neurobiología Comparada, Instituto Cavanilles, Universidad de Valencia, Valencia 46980, Spain
| | - Kari Alitalo
- Molecular/Cancer Biology Laboratory, Biomedicum Helsinki, University of Helsinki, Helsinki FI-00290, Finland
- Department of Pathology, Haartman Institute, University of Helsinki, Helsinki FI-00290, Finland
| | - Anne Eichmann
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, Paris 75005, France
- Department of Cardiology, Yale University School of Medicine, Connecticut 06511, USA
| | - Jean-Léon Thomas
- University Pierre et Marie Curie-Paris 6, Centre de Recherche de l'Institut du Cerveau et de la Moelle Epiniere, UMR S975, Paris 75651, France
- Inserm, U975, Paris 75651, France
- CNRS, UMR 7225, Paris 75651, France
- Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris 75651, France
- Department of Neurology, Yale University School of Medicine, Connecticut 06511, USA
| |
Collapse
|
50
|
Matrix-binding vascular endothelial growth factor (VEGF) isoforms guide granule cell migration in the cerebellum via VEGF receptor Flk1. J Neurosci 2010; 30:15052-66. [PMID: 21068311 DOI: 10.1523/jneurosci.0477-10.2010] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) regulates angiogenesis, but also has important, yet poorly characterized roles in neuronal wiring. Using several genetic and in vitro approaches, we discovered a novel role for VEGF in the control of cerebellar granule cell (GC) migration from the external granule cell layer (EGL) toward the Purkinje cell layer (PCL). GCs express the VEGF receptor Flk1, and are chemoattracted by VEGF, whose levels are higher in the PCL than EGL. Lowering VEGF levels in mice in vivo or ectopic VEGF expression in the EGL ex vivo perturbs GC migration. Using GC-specific Flk1 knock-out mice, we provide for the first time in vivo evidence for a direct chemoattractive effect of VEGF on neurons via Flk1 signaling. Finally, using knock-in mice expressing single VEGF isoforms, we show that pericellular deposition of matrix-bound VEGF isoforms around PC dendrites is necessary for proper GC migration in vivo. These findings identify a previously unknown role for VEGF in neuronal migration.
Collapse
|